preliminary assessment of the effect of plant...

148
PRELIMINARY ASSESSMENT OF THE EFFECT OF PLANT PROTEASE INHIBITORS ON THE EXPRESSION OF HETEROLOGOUS GENES IN PLANT CELLS NASSIM KHALIGHI FACULTY OF SCIENCE UNIVERSITI OF MALAYA KUALA LUMPUR 2011

Upload: phamkhue

Post on 26-Apr-2019

213 views

Category:

Documents


0 download

TRANSCRIPT

PRELIMINARY ASSESSMENT OF THE EFFECT OF PLANT

PROTEASE INHIBITORS ON THE EXPRESSION OF

HETEROLOGOUS GENES IN PLANT CELLS

NASSIM KHALIGHI

FACULTY OF SCIENCE

UNIVERSITI OF MALAYA

KUALA LUMPUR

2011

i

PRELIMINARY ASSESSMENT OF THE EFFECT OF PLANT

PROTEASE INHIBITORS ON THE EXPRESSION OF

HETEROLOGOUS GENES IN PLANT CELLS

NASSIM KHALIGHI

DISSERTATION SUBMITTED IN FULFILMENT OF

THE REQUIREMENT FOR THE DEGREE OF

MASTER OF BIOTECHNOLOGY

INSTITUTE OF BIOLOGICAL SCIENCES

FACULTY OF SCIENCE

UNIVERSITI OF MALAYA

KUALA LUMPUR

2011

ii

UNIVERSITI MALAYA

ORIGINAL LITERARY WORK DECLARATION

Name of Candidate: NASSIM KHALIGHI (I.C/Passport No: E12567019)

Registration/Matric No: SGF080024

Name of Degree: Master of Biotechnology

Title of Project Paper/Research Report/Dissertation/Thesis (“this Work”):

Title

PRELIMINARY ASSESSMENT OF THE EFFECT OF PLANT PROTEASE INHIBITORS ON THE

EXPRESSION OF HETEROLOGOUS GENES IN PLANT CELLS

Field of Study: PLANT MOLECULAR BIOLOGY

I do solemnly and sincerely declare that:

(1) I am the sole author/writer of this Work;

(2) This Work is original;

(3) Any use of any work in which copyright exists was done by way of fair dealing and for

permitted purposes and any excerpt or extract from, or reference to or reproduction of any

copyright work has been disclosed expressly and sufficiently and the title of the Work and its

authorship have been acknowledged in this Work;

(4) I do not have any actual knowledge nor do I ought reasonably to know that the making of this

work constitutes an infringement of any copyright work;

(5) I hereby assign all and every rights in the copyright to this Work to the University of Malaya

(“UM”), who henceforth shall be owner of the copyright in this Work and that any reproduction

or use in any form or by any means whatsoever is prohibited without the written consent of UM

having been first had and obtained;

(6) I am fully aware that if in the course of making this Work I have infringed any copyright

whether intentionally or otherwise, I may be subject to legal action or any other action as may be

determined by UM.

Candidate’s Signature Date

Subscribed and solemnly declared before,

Witness’s Signature Date

Name:

Designation:

iii

ABSTRACT

Despite the successful development of pant-based expression systems for the production

of a wide range of recombinant proteins in recent years, there remain serious limitations

in the final yield accumulation of the foreign protein in the plant cells used. Plant

protease activity represents a significant barrier to efficient and hence economical

recombinant protein production. Inadequate information is available on specific plant

proteases and its role in foreign protein degradation. Some strategies have been reported

recently to overcome endogenous plant protease effects on foreign protein integrity and

activity in plants including the co-expression of recombinant proteases to reduce the

endogenous protease activity. The lack of information in this area suggests that more

studies should be carried out to assess different plant protease inhibitors for the

improvement of the stability of foreign protein production in particular host plants. The

aim of this research was to evaluate the effect of recombinant plant protease inhibitors

on the expression of a cloned heterologous ScFv antibody gene in plant cells. The

recombinant tomato Cathepsin D Inhibitor (CDI) gene fused to a KDEL sequence was

ligated to a previously construcuted recombinant antibody (scFv) gene developed

against toxoplasmosis epitopes (pCTOXO-CDI-KDEL) and then transferred

successfully into Nicotiana tabacum cv. BY-2 cell suspension using Agrobacterium-

mediated transformation method. In addition, the scFv gene was also ligated to a

construct containing only the endoplasmic reticulum retention signal, KDEL,

(pCTOXO-KDEL) and this was also transferred into tobacco cells as a comparison

against CDI-KDEL containing cells. Both constructs contained GUS marker gene. To

assess the effect of the CDI gene in putative transformed tobacco cells GUS

fluorometric assay was performed. Total soluble protein of the tobacco cells was also

analyzed by Bradford protein assay. The tobacco cells carrying the CDI protease

inhibitor gene showed higher TSP levels in comparison with plant cells lacking this

iv

gene. This finding suggests the positive effect of co-expression of the CDI gene on the

expression of scFv anti-toxoplasmosis in tobacco cells. However, quantitative GUS

enzymatic activity expressed in pmol 4-MU/mg protein/min did not show any

significant differences between two constructs in paired t-test (p>0.05). Overall results

suggest that this strategy could be viable for increasing the production of ScFv

antibodies in plant cells.

v

ABSTRAK

Walaupun perkembangan sistem ekspresi berasaskan tumbuhan dalam penghasilan

protin rekombinan beberapa tahun kebelakangan ini telah berjaya,tetapi masih terdapat

halangan untuk memastikan pengumpulan produk akhir yang maksima di dalam sel

tumbuhan yang digunakan. Di antara faktor penghalang yang utama dalam penghasilan

protein rekombinan yang cekap dan produktif adalah aktiviti enzim protease. Walaupun

fungsi sesetengah protein protease dalam degradasi protein asing telah diketahui, namun

maklumat yang ada tidak mencukupi. Beberapa strategi dilaporkan telah digunakan

untuk mengatasi masalah ini termasuklah ekspresi bersama(co-expression) protease

rekombinan untuk mengurangkan aktiviti protease dalaman (endogenous) tumbuhan.

Kurangnya maklumat dalam bidang ini menunjukkan lebih banyak kajian perlu

dijalankan untuk menilai potensi perencat protease tumbuhan (plant protease inhibitor)

yang berbeza bagi meningkatkan pengeluaran/penghasilan protein asing dalam

tumbuhan. Kajian ini bertujuan untuk menilai kesan perencat (inhibitor) rekombinan

protease tumbuhan ke atas pengekspresan klon gen ‘heterologous ScFV’ antibodi dalam

sel tumbuhan. Gabungan antara gen ‘Cathepsin D Inhibitor’ (CDI) daripada tomato

rekombinan dan jujukan KDEL telah dicantumkan ke dalam gen rekombinan antibodi

yang dapat mengecam ‘toxoplasmosis epitope’ (pCTOXO-CDI-KDEL). Fragmen DNA

ini (pCTOXO-CDI-KDEL) seterusnya dipindahkan ke dalam cell suspension Nicotiana

tabacum cv. BY-2 melalui kaedah transformasi berasaskan Agrobacterium. Di samping

itu, gen scFv turut digabungkan ke dalam konstruk (fragmen DNA) yang hanya

mengandungi isyarat pengekalan reticulum endoplasmik, KDEL, (pCTOXO-KDEL)

dan seterusnya dipindahkan ke dalam sel tembakau untuk dijadikan perbandingan

dengan sel yang mengandungi CDI-KDEL. Kedua-dua fragmen DNA ini mempunyai

gen penanda GUS. Ujian fluorometrik GUS dilakukan untuk menganalisa/menilai kesan

gen CDI dalam sel tembakau tersebut. Jumlah protein larut dalam sel tembakau turut

vi

dianalisa menerusi ujian protin Bradford. Sel tembakau yang mengandungi gen ‘CDI

protese inhibitor’ menunjukkan jumlah protein larut yang lebih tinggi berbanding sel

tumbuhan yang tidak mempunyai gen ini. Penemuan ini menunjukkan kemungkinan

wujudnya kesan positif ekspresi bersama gen CDI ke atas pengekspresan scFv anti-

toxoplasmosis dalam sel tembakau. Namun begitu, aktiviti kuantitatif pmol enzim GUS

yang dikira berdasarkan t-berpasangan (p>0.05) dalam unit pmol 4-MU/mg protein/min

di antara kedua-dua DNA tidak menunjukkan perbezaan yang ketara. Secara

keseluruhannya, data daripada eksperimen yang dijalankan menunjukkan petanda

positif keberkesanan kaedah ini dalam meningkatkan pengeluaran antibodi scFv dalam

sel tumbuhan.

vii

ACKNOWLEDGMENT

I would like to take this opportunity to express my sincere appreciation and gratitude to

my supervisor, Prof. Rofina Yasmin Othman for her guidance and support throughout

the course of my research. Particularly, her suggestions, constructive criticisms and

encouragements have been of great value to me not only toward the writing of this

research but also during the course of the study.

Very special thanks to Dr. Yusmin Yusuf for giving invaluable advice and information

which has been of great help in the completion of my project.

This research would not have been completed without the encouragement, patience and

support of my husband, Soleyman Paydar, and not forgetting my parents.

Lastly, I would like to extend my grateful thanks to everyone who has helped me

directly and indirectly in the completion of this study.

viii

TABLE OF CONTENTS

TITLE PAGE……………………………………………………….………...…………i

ORIGINAL LITERARY WORK DECLARATION …………………....…………..ii

ABSTRACT……………………………………………………………...…………….iii

ACKNOWLEDGMENT……………………………………………..……….……....vii

TABLE OF CONTENTS……………………………………………….…..…..……viii

LIST OF FIGURES…………………………………………………….….…...……xiii

LIST OF TABLES………………………………………….….….…………………..xv

ABBREVIATION…………………………………………………………..……...…xvi

CHAPTER 1: INTRODUCTION ................................................................ 1

CHAPTER 2: LITERATURE REVIEW ..................................................... 4

2.1 PRODUCTION OF ANTIBODIES IN VITRO ............................................... 4

2.2 COMPARING DIFFERENT EXPRESSION SYSTEMS ............................... 4

2.2.1 Using plants as an expression system for protein production .................... 6

2.3 FOREIGN PROTEIN ACCUMULATION LEVEL IN PLANTS ................... 9

2.4 PROTEOLYTIC ENZYME ............................................................................. 10

2.5 MINIMIZING PROTEOLYSIS OF HETEROLOGOUS PROTEINS IN

PLANT CELLS AND TISSUES ................................................................................ 12

2.5.1 Targeting of recombinant gene expression in specific plant tissue .......... 12

2.5.2 Targeting of recombinant protein to specific organelle in plant cells ...... 14

2.5.3 Co-expression of protease inhibitors ........................................................ 17

ix

2.6 INTRODUCTION TO TOXOPLASMOSIS AND SCFV GENE .................. 19

2.7 AGROBACTERIUM MEDIATED TRANSFORMATION ........................... 20

2.8 BINARY VECTORS FOR AGROBACTERIUM MEDIATED

TRANSFORMATION ................................................................................................ 21

2.8.1 pCAMBIA Vector ...................................................................................... 21

CHAPTER 3: MATERIALS AND METHODS ....................................... 23

3.1 MATERIALS ................................................................................................... 23

3.1.1 Preparation of plasmid pCANTAB5E and pCAMBIA 1301 ..................... 25

3.1.2 Preparation of plasmid pUC-PI ................................................................ 27

3.1.3 Preparation of plasmid construct pCTOXO-KDEL .................................. 29

3.2 POLYMERASE CHAIN REACTION ............................................................ 29

3.2.1 Colony PCR for pUC-PIs .......................................................................... 29

3.2.2 PCR Confirmation for extracted pUC-PI ................................................. 30

3.2.3 PCR Confirmation for extracted pCTOXO-KDEL ................................... 31

3.2.4 PCR for anti-toxoplasmosis scFv gene ..................................................... 31

3.2.5 PCR for Protease Inhibitor genes (PIs) .................................................... 32

3.3 RESTRICTION ENDONUCLEASE DIGESTION ....................................... 33

3.3.1 Double RE digestion for anti-toxoplasmosis scFv gene ........................... 33

3.3.2 Double RE digestion for Protease Inhibitor genes ................................... 33

3.3.3 Double RE digestion for cloning vector pCAMBIA 1301 ......................... 33

3.3.4 GEL Extraction ......................................................................................... 34

3.4 CONSTRUCTION OF PCAMBIA EXPRESSION VECTORS .................... 34

x

3.4.1 pCTOXO-BBTI construct ......................................................................... 34

3.4.2 pCTOXO-CDI construct ........................................................................... 36

3.4.3 pCTOXO-OCPI construct ......................................................................... 37

3.5 COLONY PCR FOR PCTOXO-PIS CONSTRUCTS .................................... 38

3.6 CONSTRUCTION OF A SECOND PCTOXO-CDI-KDEL CONSTRUCT . 39

3.7 TRANSFORMATION OF CONSTRUCTS INTO A. TUMEFACIENS ...... 41

3.7.1 Preparation of Agrobacterium competent cell .......................................... 41

3.7.2 Agrobacterium heat shock transformation ............................................... 42

3.7.3 Colony PCR for transformed Agrobacterium with plasmid DNA constructs

42

3.8 PLANTS CELL SUSPENSION ...................................................................... 44

3.8.1 Tobacco cell suspension media preparation ............................................. 44

3.8.2 Subculturing of Tobacco cell suspension .................................................. 45

3.9 PREPARATION OF PLANT CELL SUSPENSIONS .................................. 45

3.10 PREPARATION OF AGROBACTERIUM CULTURE ................................ 46

3.11 AGROBACTERIUM-MEDIATED TRANSFORMATION OF PLANT

CELLS ......................................................................................................................... 47

3.11.1 Co-cultivation procedure .......................................................................... 47

3.11.2 Killing Agrobacterium .............................................................................. 48

3.12 ASSAY OF ß-GLUCURONIDASE (GUS) QUANTITATIVE ACTIVITY BY

FLUOROMETRY ....................................................................................................... 48

3.12.1 Preparation of sample, extraction and fluorescence 4-MU quantification

49

xi

3.12.2 Bradford protein determination ................................................................ 51

CHAPTER 4: RESULTS ........................................................................... 55

4.1 PLASMID PREPARATION ........................................................................... 55

4.1.1 Preparation of plasmid pCANTAB5E and pCAMBIA 1301 ..................... 55

4.1.2 Preparation of plasmid pUC-PI ................................................................ 56

4.1.3 Preparation of plasmid construct pCTOXO-KDEL .................................. 60

4.2 PCR FOR ANTI-TOXOPLASMOSIS SCFV AND PROTEASE INHIBITOR

GENES ........................................................................................................................ 62

4.3 DOUBLE RE DIGESTED GENES ................................................................ 63

4.4 CONSTRUCT ANALYSIS .............................................................................. 65

4.4.1 Analysis of pCTOXO-PIs .......................................................................... 65

4.4.2 pCTOXO-CDI-KDEL ................................................................................ 71

4.5 PCR ANALYSIS OF TRANSFORMED AGROBACTERIUM LBA4404

WITH PLASMID CONSTRUCTS ............................................................................. 72

4.6 TARGET SYSTEM .......................................................................................... 74

4.6.1 Tobacco cell suspension culture ............................................................... 74

4.7 QUANTITATIVE GUS FLUOROMETRIC ASSAY ..................................... 76

4.7.1 Bradford assay .......................................................................................... 79

4.8 COMPARISON OF ENZYME ACTIVITY .................................................... 81

CHAPTER 5: DISCUSSION ..................................................................... 84

5.1 PLASMID CONSTRUCT DEVELOPMENT ................................................ 85

5.1.1 Verification of Ligation ............................................................................. 85

xii

5.2 COMPARISON OF TOTAL SOLUBLE PROTEIN ..................................... 87

5.3 COMPARISON OF ENZYME ACTIVITY .................................................... 88

CHAPTER 6: CONCLUSION ................................................................... 90

6.1 FUTURE STUDIES ........................................................................................ 91

REFERENCE: ........................................................................................... 92

APPENDICES .......................................................................................... 101

APPENDIX - A ......................................................................................................... 102

APPENDIX - B ......................................................................................................... 114

APPENDIX - C ......................................................................................................... 115

APPENDIX - D ......................................................................................................... 116

APPENDIX - E ......................................................................................................... 117

APPENDIX - F ......................................................................................................... 119

APPENDIX - G ......................................................................................................... 120

APPENDIX - H ........................................................................................................ 121

APPENDIX - I .......................................................................................................... 122

APPENDIX - J ......................................................................................................... 123

APPENDIX - K ......................................................................................................... 124

APPENDIX - L ......................................................................................................... 125

APPENDIX - M ........................................................................................................ 126

xiii

LIST OF FIGURES

Figure 3.1: pCTOXO-PI construct

Figure 3.2: General Overview of experiments conducted in this research

Figure 3.3: pCTOXO-BBTI construct

Figure 3.4: pCTOXO-CDI construct

Figure 3.5: pCTOXO-OCPI construct

Figure 3.6: Ligation product of pCTOXO-CDI-KDEL

Figure 3.7: pCTOXO-CDI-KDEL construct

Figure 4.1: Plasmid preparations for pCANTAB5E carrying scFv genes

Figure 4.2: Plasmid preparations for pCAMBIA 1301

Figure 4.3: Colony PCR for pUC-PIs carrying PI genes transformed into JM109

Escherichia coli competent cells.

Figure 4.4: Plasmid preparation for pUC57 vector carrying PI genes (pUC-PI) from

transformed Escherichia coli.

Figure 4.5: PCR verification for plasmid pUC57 carrying PI genes after plasmid

preparation.

Figure 4.6: Plasmid preparation for pCTOXO-KDEL plasmid construct.

Figure 4.7: PCR product for pCTOXO-KDEL using scFv-KDEL primers.

Figure 4.8: Purified PCR product of anti-toxoplasmosis scFv genes using scFv primers.

Figure 4.9: Purified PCR product of PI genes using PI primers.

xiv

Figure 4.10: Purified RE digested scFv genes and pCAMBIA 1301

Figure 4.11: Purified RE digested Protease Inhibitor genes

Figure 4.12: Colony PCR for pCTOXO-PIs using scFv primers.

scFv15+PIs+pCAMBIA1301

Figure 4.13: Colony PCR for pCTOXO-PIs using scFv primers.

scFv9+BBTI+pCAMBIA1301

Figure 4.14: Colony PCR for pCTOXO-BBTI using scFv-PI flanking primers.

Figure 4.15: Colony PCR for pCTOXO-CDI using scFv-PI flanking primers.

Figure 4.16: Colony PCR for pCTOXO-OCPI using scFv-PI flanking primers.

Figure 4.17: PCR for ligated plasmid construct pCTOXO- CDI-KDEL using new scFv-

PI primers.

Figure 4.18: Colony PCR for Agrobacterium transformed with pCTOXO- CDI-KDEL

plasmid construct.

Figure 4.19: Colony PCR for Agrobacterium transformed with pCTOXO-KDEL

plasmid construct.

Figure 4.20: Nicotiana tabacum cv. BY-2 cell suspension

Figure 4.21: Comparison of TSP of tobacco cells transformed with pCTOXO- CDI-

KDEL (A1 & A2 colonies) and pCTOXO-KDEL (B1 & B2 colonies).

Figure 4.22: Comparison of the level of GUS Activity of the transformed tobacco cells

with two different plasmid constructs

Figure 5.1: Matching of compatible sticky ends

xv

LIST OF TABLES

Table 3.1: 4-MU Standard Dilutions

Table 3.2: Bradford Standard Measurements

Table 4.1: Number and percentage of surviving and dead tobacco cell suspension

cultures after 14 days of co-cultivation with Agrobacterium carrying plasmid construct

pCTOXO-CDI-KDEL and pCTOXO-KDEL.

Table 4.2: The relative fluorescent intensity of test samples for three replicates of each

colony transformation A1, A2, B1, B2

Table 4.3: Relative Fluorescent Intensity of test samples (pmol)

Table 4.4: The OD values for test samples and protein conc. of samples

Table 4.5: Comparison of 4-MU enzyme activity between A1 vs. B1 and A2 vs. B2

samples. 4-MU enzyme activity expressed in pmol/mg. protein/min for tobacco cells

transformed with pCTOXO- CDI-KDEL (A1 and A2 Agrob colonies) and pCTOXO-

KDEL (B1 and B2 Agrobacterium colonies).

xvi

ABBREVIATION

A: Absorbance

Agrob: Agrobacterium

anti-toxo: anti-toxoplasmosis

BBTI: Bowman–Birk trypsin inhibitor

bp: base pair

BSA: Bovine serum albumin

BY-2: Bright Yellow–2

CaCl2: Calcium chloride

CDI: Cathepsin D Inhibitor

cDNA: Complementary deoxyribonucleic acid

conc: Concentration

cv: Caltivar

DNA: deoxyribonucleic acid

E.coli: Escherichia coli

EDTA: Ethylenediaminetetracetic acid

ER: Endoplasmic reticulum

ETOH: Ethanol

Fab: Fragment antigen binding

xvii

g: Gram

GAB: GUS assay buffer

GEB: GUS extraction buffer

HDEL: His-Asp-Glu-Leu

HIV: human immunodeficiency virus

Kb: Kilobase

KCl: Potassium chloride

KDEL: Lys-ASP-Glu-Leu

l: Liter

LB: Luria-Bertani

M: Molar

M2: Media 2

mAB: Monoclonal antibody

mg: milligram

min: minute

ml: milliliter

mM: Millimolar

MnCl2: Manganese chloride

MOPS: 3-(N-morpholino)propanesulfonic acid

MS: Murashige and Skoog media

xviii

MSO: Murashige and Skooge basal medium

MUG: 4-methylumbelliferyl-ß-D-glucuronide

MW: Molecular weight

Na2CO3: Sodium carbonate

NaOH: Sodium hydroxide

nM: Nanomolar

OCPI: Oryzacystatin protease inhibitor I

OD: Optical density

PCR: polymerase chain reaction

pCTOXO-BBTI: pCAMBIA 1301-scFv anti –Toxo-BBTI

pCTOXO-CDI-KDEL: pCAMBIA 1304-scFv anti –Toxo-CDI-KDEL

pCTOXO-CDI: pCAMBIA 1301-scFv anti –Toxo-CDI

pCTOXO-KDEL: pCAMBIA 1304-scFv anti –Toxo-KDEL

pCTOXO-OCPI: pCAMBIA 1301-scFv anti –Toxo-OCPI

pCTOXO-PI: pCAMBIA 1301-scFv anti –Toxo-Protease inhibitor

pH: Potential hydrogen

PI: Protease inhibitor

pmol: picomole

pUC-PI: pUC57-Protease inhibitor

xix

RE: Restriction endonuclease

RNA: ribonucleic acid

RNAse A: Ribonuclease A

rpm: Revolutions per minute

scFv: Single chain variable fragment

sdH2O: Sterile distilled water

SDS: Sodium dodecyl sulphate

sec: Second

TBE: Tris borate

Ti: Tumor inducing

TOXO: Toxoplasmosis

Tris-Cl: Trisbase-Hydrochloric acid

TSP: Total soluble protein

u: Unit

UV: Ultraviolet

v: Volume

Vir: Virulence

vs: Versus

w: Weight

xx

w/v: Weight over volume

X: Times

2,4-D: 2,4-Dichlorophenoxyacetic acid

4-MU: 4-methylumbelliferone

µ µl: Micro liter

g: Micro gram

µM: Micro molar

˚C: Degree Celsius

%: Percentage

1

CHAPTER 1: INTRODUCTION

The demand of producing different recombinant proteins is increasing in recent years.

For instance production of useful therapeutic recombinant proteins is highly needed in

order to improve the health level of human society. To date, several heterologous

expression systems have been designed for the production of useful therapeutic

recombinant proteins. In most cases, eukaryotic hosts competent in performing complex

posttranslational modifications have been involved. But, plant-based systems compared

favorably with alternative expression platforms, both in terms of quality and cost of

complex therapeutic proteins. Higher plants are used as versatile expression systems for

producing therapeutic foreign proteins. This technology offers unique advantages,

including cost reduction, possibility of bulk production of useful foreign proteins as

well as the range of plant hosts available and the stability of transgenic lines in seeds

that might allow export of transgenic crops (containing, for example, medicinal

compounds) to deprived areas of the world .

One major problem of producing recombinant proteins in higher plants is instability of

produced recombinant proteins. This system presents serious challenges to protein

integrity. That means foreign protein degradation directly impacts on the final yield,

homogeneity and overall quality of the resulting protein product. The instability of

recombinant proteins is due to activity of plant proteases. The proteolytic enzymes in

plant cells, in particular, are often a major hurdle to recombinant protein integrity, both

in planta at the expression stage and in vitro during protein recovery from plant tissues.

Therefore, in plant expression systems proteolytic processing, notably, may

dramatically alter the structural integrity and overall accumulation of recombinant

proteins in plant expression systems.

2

Although several strategies have been considered to minimize proteolysis in plant

protein factories involving the targeting of transgene expression or protein accumulation

to specific tissues or cellular organelles, this problem still needs a lot of researches to

overcome this negative effect on foreign protein final yield.

A recent solution proposed is the co-expression of companion protease inhibitors

interfering with endogenous proteases which are active against specific endogenous

proteases. Recombinant protease inhibitors could prove functional to modulate

proteolytic activities in situ but very few researchers conducted in this area so there is a

need to conduct more investigations to improve this area of research to help improving

the yield and functionality of foreign proteins.

The lack of sufficient information on different plant proteases existence in plants makes

conducting more researches in this area necessary. The Nicotiana tabacum plant as a

very important target system in producing foreign proteins has shown the proteolytic

degradation problem. However many of the proteases responsible for the recombinant

protein degradation in this plant remained unknown.

For obtaining the information on type of proteases which exist in tobacco plant cells one

way is to assess the co-expression of protease inhibitor genes with recombinant protein

gene. The improved level of desired foreign protein translation in plant cells carrying

the suitable companion protease inhibitor can make the production of foreign proteins

more efficient in tobacco cells as a very useful target system.The Cathepsin D inhibitor

gene, Bowman Birk Trypsin I inhibitor and Orysacystatin protease Inhibitor genes were

designed to assess the presence of proteases that can become inactive by the help of co-

expression of these inhibitors in tobacoo cells.

3

In this research, the tomato Cathepsin D Inhibitor gene was ligated to a recombinant

single chain fragment anti-toxiplasmosis antibody (-scFv). The construct was then

transferred and expressed in Nicotiana tabacum cultivar BY-2 cells. In addition, the ER

retention signal, KDEL, ligated to scFv (obtained from previous studies) was transferred

and expressed in tobacco cell suspension in separate experiments to produce putatively

transformed tobacco plant cells carrying scFv gene but lacking CDI protease inhibitor.

The expression of scFv in the presence of the CDI protease inhibitor in plant cells was

investigated and compared to heterologous scFv gene expression in plant cells without

CDI protease inhibitor.

The objective of this research was to assess the effect of plant protease inhibitors on the

expression of heterologous monoclonal antibody scFv gene of anti-toxoplasmosis in

plant cell suspension.

The specific objectives are to develop constructs of scFv-PIs, carry out transformation

experiments and then analyze the resulting putative transformed plant cells in terms of

total soluble protein (TSP) and GUS Enzymatic activity using Bradford protein assay

and GUS Fluorometric Assay.

4

CHAPTER 2: LITERATURE REVIEW

2.1 PRODUCTION OF ANTIBODIES IN VITRO

Today antibodies are applied in the diagnosis of diseases and therapeutic uses, by-

research institutions and pharmaceutical companies (Joosten et al., 2003). The

production of antibodies have increased in quality and quantity in recent years by the

help of recombinant DNA technology, so the expression of a whole antibody gene or a

fragment of it has become achievable in different living organisms’ cells such as

bacteria (Nouaille et al., 2003), mammalian cells (Yazaki et al., 2004) and plant cells.

2.2 COMPARING DIFFERENT EXPRESSION SYSTEMS

The production of heterologous proteins is mostly carried out in prokaryotic systems

such as cultures of bacterial cells (Lu Yinghua et al., 2004). In many cases the

prokaryotic system is preferred because the bacterial cells can be grown rapidly and

high cell densities can be achieved easily, besides the media in this system is cheap (Lu

Yinghua et al., 2004). However, there are some limitations to use of the prokaryotic

systems. Firstly, the lack of post-transcriptional modifications such as phosphorylation,

acylation, and N-and O-linked glycosylation which can affect the recombinant protein

activity (Lu Yinghua et al., 2004), since the ability to perform post translational

modifications does not exist in bacterial expression systems (Jung and Williams, 1997).

Secondly, the heterologous proteins may accumulate into insoluble structures because

the prokaryotic expression system does not have the ability to secrete the protein

product into the extracellular medium, and proteins may remain in structures known as

5

inclusion bodies. Therefore, the downstream processing of heterologous proteins

become very expensive and complicated (Lu Yinghua et al., 2004).

The eukaryotic system was then established to overcome the problem of the prokaryotic

system. One of the popular eukaryotic systems is yeast. The proteins produced in yeast

system can be secreted to the media. The viruses that can affect humans do not exist in

yeasts (Lu Yinghua et al., 2004). Yeast are able to glycosylate the proteins that are

produced in their system and they can multiply rapidly. However, the folding of

manufactured protein is different from the type seen in mammalian cell cultures. The

difference is glycosylation process. Hyper mannosylation is another disadvantage of

yeast system. It means that proteins produced in yeast normally contain extra mannose

added to the basic oligosaccaharide and thus such manufactured proteins are not usually

appropriate for the production of human medicines (Jung and Williams, 1997).

A higher eukaryotic system in protein production process in comparison with yeast cells

are insect cells. This system also has been used for the production of useful proteins. An

example of such expression system is the baculovirus (Rai and Padh, 2001) but, with

some limitations. In baculovirus expression system the proteolytic cleavages in lysine or

arginine rich sequences is ineffective. This deficiency causes imperfect protein folding.

Other disadvantages of this system are expensive media for the production of insect

cells and slow growth of the cells (Lu Yinghua et al., 2004).

The other eukaryotic system in protein production process is mammalian cells. The

mammalian cells are generally used for high value end products. Performing the most

suitable type of post-translational modifications to the protein product is one of the

advantages using this expression system. This benefit of the mammalian cellular

6

expression system makes them appropriate for use in human medication (Rai and Padh,

2001). However, there are some problems associated with the use of this mammalian

cell system. This expression system is very hard to maintain and it is a time consuming

system. The production and maintenance of this system is a very complex task in

comparison with the other biological expression systems and the yield of the protein

product is normally low, so only when other systems are incompatible mammalian cells

are used as the expression system (Rai and Padh, 2001).

2.2.1 Using plants as an expression system for protein production

An attractive host system for production of recombinant proteins such as

biopharmaceuticals is plant-based expression system. Plants offer several important

advantages over other heterologous expression systems. As a eukaryote system, plants

are able to fold and assemble complex proteins correctly and post-translational

modifications like glycosylation can be achieved in plants. The requirement for animal

cells, animal derived culture materials or potentially infectious animals is also

eliminated with applying the plant host system in the production of foreign proteins. An

alternative technique for the production of recombinant proteins is plant cell culture in

bioreactors. The control of production environment is easier in plant cultures in

comparison with whole plants. Besides, the process cycle is shorter and purification

process is simpler and cheaper due to secretion of proteins from plant cells (Doran,

2006).

The first successful animal antibody production was achieved in transgenic tobacco

plants in the late 1980’s (Hiatt et al., 1989). That successful experiment showed

recombinant monoclonal antibodies can be produced in plants as an intact antibody

7

molecule. In that work cDNAs encoding either the heavy chain or light chain of a

catalytic mouse antibody molecule were expressed in the engineered plants. Then, the

plants were cross pollinated. As a result both cDNAs were found to be expressed in the

progeny, and the fully assembled recombinant antibody expressed in plants. Almost 1%

of total soluble protein (TSP) in the plant was found to be comprised of the recombinant

antibody. The antibody molecules produced in plant tissue had the same antigen-

binding activity like the progenitor antibody. Since then, expression of antibody

molecules in plants has been done by several groups for either modification or

improvement of plant characteristics and performance or for large-scale production of

antibodies using plants as bioreactors (Ma et al., 1995). The techniques for the

production of human recombinant proteins in plants have improved a lot and the

techniques have become more efficient. In recent years, the production of monoclonal

antibodies both in the form of full length or antibody fragments preferably has been

done on plant system by the scientific community (Hendy et al,. 1999) instead of using

traditional microbial systems. The large scale production of commercially important

biomolecules can be achieved in plants (Whitelam and Cockburn, 1997).

In general, plant expression systems produce recombinant biomolecules more

efficiently, both in terms of quality and cost of complex therapeutic proteins in

comparison with other expression platforms (Komarnytsky et al., 2006). The range of

plant hosts available and the possibility of bulk production of recombinant therapeutic

proteins and reducing costs are some of the advantages of using plant-based technology.

The export of transgenic plants to deprived areas of the world is also another advantage

of this system. Using stable transgenic lines in seeds containing medicinal compounds is

an example for this benefit (Ma et al., 1995). The heterologous production of clinically

8

useful proteins as therapeutic agents can be achieved in higher plants which represent

versatile expression platforms.

2.2.1.1 Plant cell cultures expression systems

Culturing the specific organs or cells that produce the antibody and either isolate the

antibody from these cells or tissues, or collect it from the culture medium is an

alternative system to whole plants expression systems. A number of different culture

systems have been developed, although most research has focused on cell suspension

cultures. Suspension cells are individual plant cells and small aggregates thereof

growing in liquid medium in a fermenter (Hellwig et al. 2004; Doran 2006). Suspension

cell cultures are usually derived from callus tissue by the disaggregation of friable callus

pieces in shake bottles, and are later scaled up for fermenter-based production.

Recombinant antibody production is achieved by using transgenic explants such as

Nicotiana tabacum to derive the plant cell cultures, or transforming the cells after

disaggregation, usually by co-cultivation with Agrobacterium tumefaciens (Twyman et

al., 2007).

Many foreign proteins have been expressed successfully in suspension cells, including

antibodies, enzymes, cytokines and hormones (Hellwig et al. 2004, Fischer et al. 1999).

Tobacco cultivar Bright Yellow 2 (BY-2) is the most popular source of suspension cells

for molecular farming, since these proliferate rapidly and are easy to transform (Nagata

et al., 1992). Other than tobacco, rice suspension cells have also been used to produce

several antibodies (Torres et al., 1999). Recombinant antibodies expressed in plant cell

suspension cultures may be secreted into the culture supernatant or retained within the

cells. Localization depends on expression construct design and the permeability of the

plant cell wall to the antibody.

9

The inclusion of a C-terminal KDEL sequence results in higher levels of antibody

accumulation in cultured cells because the biochemical environment of the endoplasmic

reticulum favors stable protein folding and assembly while reducing the level of

proteolytic degradation. However, this also makes it necessary to disrupt the cells in

order to isolate the protein, which requires additional processing time and causes the

release of phenolic molecules that interfere with purification and reduce production

yield. Thus, the preferred approach is to secrete the target proteins and capture them

from the culture supernatant or release them from the cells by mild enzymatic digestion.

(Twyman et al., 2007).

2.3 FOREIGN PROTEIN ACCUMULATION LEVEL IN PLANTS

The accumulation level of foreign protein product in plant system is a very challenging

and crucial parameter which affects the economics of protein production. A significant

problem that limits the commercial exploitation of recombinant plant as host system is

low protein yield (Doran, 2006). The accumulation level of most foreign proteins is

much lower than 1% of total soluble protein (TSP) in plant biomass; Commonly, about

0.01–0.1% TSP or less are reported as maximum product concentrations ( Daniell et al.,

2001). The low yield of recombinant protein is usually considered inadequate for

competition with other heterologous protein production systems and it remains as a

major difficulty in this technology (Kusnadi et al., 1997). Therefore, the future

development of this technique is directly affected with improving foreign protein

accumulation in plant systems.

In recent years, there is an increasing recognition that shows overall yield of foreign

proteins in plant cells are crucially determined by protein degradation. At any given

time, the amount of recombinant protein found in plant cells expresses a balance

10

between protein production and protein degradation or loss. As evidence, several

recombinant plant systems have been reported in which there is a lack of correlation

between foreign mRNA transcript levels and foreign protein product concentration

(Richteret al., 2000; Outchkourov et al., 2003). It suggests that foreign protein may be

expressed successfully but subsequently degraded. Therefore, high mRNA levels do not

guarantee high accumulation levels of foreign protein (Doran, 2006).

2.4 PROTEOLYTIC ENZYME

Higher plants are used as versatile expression systems for producing therapeutic foreign

proteins. However, this system presents serious challenges to protein integrity. The

instability of recombinant proteins is due to activity of plant proteases. The well-

documented importance of proteolytic enzymes in plant cells, in particular, is often a

major hurdle to recombinant protein integrity, both in planta at the expression stage and

in vitro during protein recovery from plant tissues (Faye et al., 2005). Foreign protein

degradation in plant expression systems directly affects the final yield, homogeneity and

overall quality of the resulting protein product. Proteolytic processing may dramatically

change the overall accumulation and structural integrity of foreign proteins in plant-

based expression systems (Benchabane et al., 2008).

Proteolytic enzymes, or proteases, contribute to the overall control of metabolic and

transduction pathways by directing the activation or hydrolysis of proteins implicated in

key regulatory processes, or by contributing to the elimination of misfolded proteins and

the selective recycling of amino acids from short-lived proteins (Vierstra, 2003;

Schaller, 2004). For the efficient processing and quality control of proteins, proteolytic

11

enzymes are essential and are a fundamental element in the reaction of cells to changing

environmental conditions (Doran , 2006).

A significant challenge for producing foreign proteins economically in all heterologous

systems, including plants, is countering the effects of protease activity. Loss of

recombinant protein after synthesizing, assembly and, in some cases, glycosylation and

post-translational modification processes represents a considerable waste of

biosynthetic resources in transgenic plant cells. Increasing protein product heterogeneity

and lower consistency are other detrimental consequences of proteolysis that can cause

regulatory problems. Therefore, more difficult and expensive downstream processing is

needed due to the presence of incomplete protein or protein fragments (Stein et al.,

2001).

Degrading abnormal or incorrectly processed proteins is an important role of proteases.

Therefore, the susceptibility of recombinant proteins to protease attack in plant cells

could reflect their improper synthesis or assembly. Folding and quaternary structure of

foreign proteins are affected by differences between plant and animal glycans. This

issue can make plant-derived proteins more susceptible to protease activity than their

animal-derived counterparts (Doran, 2006). Misfolding and lack of proper disulphide

crosslinking in plant-derived proteins can also increase the likelihood of protease attack

(Smith et al., 2002).

12

2.5 MINIMIZING PROTEOLYSIS OF HETEROLOGOUS PROTEINS IN

PLANT CELLS AND TISSUES

Specific organelles or sub-cellular locations within plant cells and tissues may be

associated with proteolytic degradation of recombinant proteins. Plant vacuoles were

identified as a possible site of foreign protein degradation in Arabidopsis thaliana

(Yang et al., 2005) and potato leaves (Outchkourov et al., 2003). Vacuoles contain a

variety of proteases that are active under mildly acidic conditions. Particular cleavage of

glycosylated antibody in tobacco cells were identified to occur in the secretory pathway

between the ER and golgi. The proteases were considered most likely to be responsible

for the cleavage (Sharp et al., 2001). Besides, the production of proteolytic fragments in

several transgenic plant systems has been reported in apoplast or plant cell wall

(Outchkourov et al., 2003; Sharp et al., 2001; Stevens et al., 2000).

2.5.1 Targeting of recombinant gene expression in specific plant tissue

The final yield of foreign protein product and its quality are strongly influenced by

specific tissue in which foreign protein is produced. Because the quantity and quality

(or overall substrate specificity) of plant proteases, notably, differ from one tissue to

another (Schaller, 2004), with a possible differential impact on the integrity of proteins.

An accumulation site with low levels of overall proteolytic activity or with endogenous

protease enzymes with no or little specific activity against accessible peptide bonds in

the protein of interest is desirable in effective foreign protein production.

To date, green leaves have been the destination of choice for several foreign proteins,

given their rapid growth rate, the possibility in some platforms to harvest leaf material

13

more than once over the growing season, and the availability of numerous regulatory

sequences well adapted to transgene expression in the leaf cell environment (Daniell et

al ., 2001). However, high protein synthesis and turnover rates which are due to the

highly active metabolism of leaf tissues may represent a significant hurdle to protein

accumulation in vivo. In particular, increased protease levels in senescing leaves (Kato

et al ., 2004, 2005; Lin and Wu, 2004; Otegui et al ., 2005; Parrott et al ., 2005)

represent a potential drawback in leaf-based production systems (Stevens et al .,2000;

Birch-Machin et al ., 2004).

By contrast with leaf tissues, efficient deposition and post-harvest storage of foreign

proteins have been reported in the seeds and storage organs of several crops. In general,

seeds appear to be a suitable tissue for recombinant protein production (Stoger et al.,

2005). The low abundance of active proteases in seed tissues during dormancy, together

with the, desiccated nature of mature seeds prevent extensive proteolysis and promote

long-term stability of proteins in planta (Fiedler and Conrad, 1995; Stoger et al., 2000).

Seeds have the appropriate molecular environment to promote protein accumulation,

and achieve this through the creation of specialized storage compartments such as

protein bodies and storage vacuoles that are derived from the secretory pathway.

Antibodies expressed in seeds remain stable for at least three years at ambient

temperatures with no detectable loss of activity (Stoger et al., 2005)). Similarly, the

storage roots and tubers of some plants show reduced metabolic activity, and may

represent an interesting solution for foreign protein storage (Artsaenko et al., 1998).

14

2.5.2 Targeting of recombinant protein to specific organelle in plant cells

Organelles have specific, complementary functions in the cell and therefore harbour

their own metabolic machinery, including a protease complement well adapted to their

specific enzymatic and physicochemical environment (Callis, 1995). Not surprisingly,

the recombinant proteins accumulation rate is strongly influenced by targeting of

foreign protein to different organelles using appropriate peptide signals. In practice, the

choice of a suitable cellular destination will also depend on the structural characteristics

of the foreign protein, which will often dictate specific co- or post-translational

modifications essential for adequate activity, stability and/or homogeneity (Faye et al.,

2005).

2.5.2.1 Targeting and retention of recombinant protein in the endoplasmic

reticulum

Proteins carrying a signal peptide for cellular secretion first enter the endoplasmic

reticulum via the ER protein translocation channel (Galili et al., 1998), and then migrate

through this compartment and the golgi apparatus until reaching the extracellular

medium (default pathway) or the vacuole, if a vacuolar sorting signal is found in the

primary sequence. Retention of foreign proteins in this compartment is also achievable

for proteins entering the ER by simple apposition of the tetrapeptide ER retention signal

(K /H)DEL (Michaud et al., 1998) at the C-terminus. The (K/H)DEL motif is a common

ER retrieval signal in eukaryotes, believed to redirect tagged proteins to the ER after

their recognition by a (K/H)DEL receptor complex in the golgi apparatus (Pagny et al.,

2000). Numerous studies have been published illustrating the positive impact of

retaining clinically or industrially useful proteins in the ER compartment of plant cells,

using, in most cases, a (K/H)DEL retention signal (Ma et al., 2003). The inclusion of a

15

C-terminal KDEL sequence results in higher levels of antibody accumulation in

cultured cells because the biochemical environment of the endoplasmic reticulum favors

stable protein folding and assembly while reducing the level of proteolytic degradation

(Twyman et al., 2007). Biologically active mAbs require a number of assembly steps

and posttranslational modifications that are carried out in the endoplasmic reticulum

(ER) (Deneke, J., 1990).

At the biochemical level, the low abundance of proteolytic enzymes and the presence of

molecular chaperones in the ER, together with an oxidizing status favouring disulphide

bond formation, make this organelle a suitable destination for several proteins

susceptible to rapid turnover or showing a complex folding pathway (Nuttall et al.,

2002; Faye et al., 2005).The ER, which constitutes a natural reservoir for some storage

proteins in seed cells (Shewry and Halford, 2002), can physically accommodate high

levels of foreign protein product in planta (Wandelt et al., 1992).

2.5.2.2 Targeting of recombinant protein to the vacuole

There are two different types of vacuole in plant cells: lytic vacuoles, which are rich in

hydrolytic enzymes, so they have an acidic environment; and protein storage vacuoles.

The second type shows a slightly acidic or neutral pH which is in favor of protein

storage (Robinson et al., 2005). Targeting to the vacuole, although not yet fully

understood, is determined by small stretches of amino acids within the protein primary

sequence acting as sorting signals to direct the maturing protein towards the vacuole

(Mackenzie, 2005; Vitale and Hinz, 2005). In general, lytic vacuoles are not considered

as a suitable destination for recombinant proteins in planta, owing to their high

proteolytic content (Goulet and Michaud, 2006). By contrast, protein storage vacuoles

16

present a milder environment compatible with protein accumulation (Stoger et al.,

2005), especially in seeds, where they are most abundant (Park et al., 2004).

2.5.2.3 Targeting of recombinant protein to the chloroplast

The chloroplast, peroxisome and nucleus have been proposed as other cellular

destinations for protein production in plant platforms (Daniell et al., 2002; Hyunjong et

al., 2006). In practice, recombinant proteins may be sent to these organelles by the

inclusion of an appropriate targeting peptide (or localization signal) in the transgene

sequence. Chloroplast transformation offers several advantages over nuclear

transformation, including uniform transgene expression rates, multiple copies of the

transgene in each cell, co-expression of multiple genes from the same construct,

minimal gene silencing and minimal transgene escape in the environment owing to the

maternal inheritance of chloroplast DNA in several species (Daniell et al., 2002). The

chloroplast stroma supports protein post translational modifications, such as

multimerization and disulphide bridge formation (Daniell, 2006), making it a suitable

environment for the expression of proteins not relying on complex modifications, such

as glycosylation, typical of the cell secretory pathway.

Recombinant protein degradation by chloroplast proteases might appear, however, to be

a non-relevant issue in terms of net production yields for some proteins expressed at

very high levels (Daniell, 2006). The proteolysis-labile protein human serum albumin,

for instance, was found at levels reaching 11% of TSP in transplastomic tobacco lines

developed using chloroplast untranslated regions in gene constructs, in sharp contrast

with levels below 0.02% of TSP in lines developed using the commonly employed

Shine–Delgarno regulatory sequence (Fernandez-San Millan et al., 2003).

17

However, there are two disadvantages to the chloroplast system – first, chloroplast

transformation is not a standard procedure and is thus far limited to a relatively small

number of crops (tobacco, tomato, potato, cotton, soybean and most recently lettuce and

cauliflower (Daniell et al. 2005b; Lelivelt et al. 2005; Nugent et al. 2006). Secondly,

they lack much of the eukaryote machinery for posttranslational modification, i.e. they

are unable to synthesize glycan chains. For this reason, they would be suitable for the

production of scFvs but not full-size immunoglobulins.

2.5.3 Co-expression of protease inhibitors

Several strategies have been considered recently to minimize proteolysis in plant protein

factories. Common strategies to overcome unwanted proteolysis in planta involve the

targeting of transgene expression or protein accumulation to specific tissues or cellular

organelles. Approaches involving the grafting of protein-stabilizing fusion domains to

recombinant proteins or the co-expression of companion protease inhibitors interfering

with endogenous proteases have also been proposed recently (Benchabane et al., 2008).

The strategy proposed recently to increase recombinant protein production in planta

involving the use of transgenic hosts with reduced proteolytic capacities is studied in

plant made pharmaceuticals. In theory, protease processes affecting recombinant protein

accumulation could be contained using antisense or RNA silencing strategies

implemented in transgenic host plants (Watson et al., 2005). Alternatively, recombinant

protease inhibitors active against specific endogenous proteases could prove functional

to modulate proteolytic activities in situ (Faye et al., 2005). Recent evidence in the

literature has suggested that the ectopic expression of protease inhibitors may have a

18

positive impact on protein levels in leaves, with negligible effects on growth and

development (Benchabane et al., 2008).

2.5.3.1 Introduction of plant protease inhibitors

Monocot plants such as tobacco, potato, tomato, banana, rice and etc, are used as

alternative systems for production of pharmaceutical proteins however there are several

proteases in these plant cells resulting in low yields of the transgenes. Some of these

proteases are known and have shown negative effects on expression of heterologous

proteins in these plants. Some are known as Trypsin, Chymotrypsin and Cystatine

proteases. The importance of trypsin and Chymotrypsin-like activities in crude protein

extracts of potato (Solanum tuberosum L.) leaves have been confirmed (Rivard et al.,

2006).

The broad-spectrum inhibitor tomato cathepsin D inhibitor (CDI), for instance, has been

shown to yield increased TSP levels (by 20%–35%) in leaves of transgenic potato lines

accumulating this inhibitor in the cytosolic compartment (Michaud et al., 2005).

Likewise, the rice cysteine protease inhibitor oryzacystatin I (OCPI) led to total protein

levels higher than expected in tobacco leaf tissues expressing this inhibitor in the

cytosol (Van der Vyver et al., 2003).Stabilized recombinant antibodies secreted by the

roots of transgenic tobacco plants also expressing (and co-secreting) a Bowman–Birk

trypsin inhibitor (BBTI) from soybean was successfully studied (Komarnytsky et al.,

2006).

19

2.6 INTRODUCTION TO TOXOPLASMOSIS AND SCFV GENE

Toxoplasmosis is a parasitic disease caused by the protozoan Toxoplasma gondii. (Ryan

KJ; Ray CG. 2004). The primary host of Toxoplasma gondii is the cat family. However,

it is able to infect most animals, including humans. Eating infected meat, ingestion of

feces of an infected cat and transmission from mother to fetus are the important ways of

infection to toxoplasmosis in animals. Although cats are often blamed for spreading

toxoplasmosis, contact with raw meat is a more significant source of human infections

in many countries, and faecal contamination of hands is a greater risk factor (Torda A

2001). The parasite can cause encephalitis (inflammation of the brain) and neurologic

diseases and can affect the heart, liver, inner ears and eyes (chorioretinitis). However,

people with a weakened immune system, such as those infected with advanced HIV

disease or those who are pregnant, may become seriously ill, and it can occasionally be

fatal.

The recombinant single chain fragment anti toxiplasmosis scFv antibody is a

recombinant antibody designed to be produced in recombinant host systems. It is

important to understand the mechanisms of action of the antibody in designing clinical

antibodies for production in heterologous expression systems. Binding to antigen is the

primary function of antibodies. Therefore, it can be sufficient to produce the antigen-

binding domain of the antibody molecule alone for the recombinant protein to be active.

The single-chain Fv molecules, which consist of the variable domains of the light and

heavy chains joined by a peptide linker and Fab fragments (fragment antigen- binding)

is an example of this approach (Ma et al., 1995).

20

2.7 AGROBACTERIUM MEDIATED TRANSFORMATION

Agrobacterium tumefaciens is a soil-borne, gram-negative bacterium. This bacteria is

the causative agent of crown gall disease, an economically important disease of a wide

range of plants. Crown-gall formation depends on the presence of a plasmid in

A.tumefaciens known as the Ti (tumour-inducing) plasmid. Part of this plasmid (the T-

DNA region) is actually transferred from the bacterium and into the plant cell, where it

becomes integrated into the genome of the host plant. The T-DNA region of any Ti

plasmid is defined by the presence of the right and left-border sequences. These border

sequences are 24-bp imperfect repeats. Any DNA between the borders will be

transferred into the genome of the host plant (Slater et al., 2003).

The T-DNA contains two types of genes: the oncogenic genes, encoding for enzymes

involved in the synthesis of auxins and cytokinins and responsible for tumour

formation; and the genes encoding for the synthesis of opines, a product resulted from

condensation between amino acids and sugars, which are produced and excreted by the

crown gall cells and consumed by A. tumefaciens as carbon and nitrogen sources.

Outside the T-DNA, are located the genes for the opine catabolism, the genes involved

in the process of T-DNA transfer from the bacterium to the plant cell (Zupan et al.,

1995). Virulent strains of A. tumefaciens contain a large megaplasmid (more than 200

kb) that plays a key role in tumour induction and for this reason it was named Ti

plasmid. The transfer is mediated by the co-operative action of proteins encoded by

genes determined in the Ti plasmid virulence region (vir genes) and in the bacterial

chromosome, chvA, chvB, psvA and att which are involved in the binding of bacteria to

the injured plant cell wall (Slater et al., 2003).The 30 kb virulence (vir) region is a

region organised in six operons that are essential for the T-DNA transfer (virA, virB,

virD, and virG) or for the increasing of transfer efficiency (virC and virE) (Zupan et al.,

21

1995; Jeon et al., 1998). The initial results of the studies on T-DNA transfer process to

plant cells demonstrate three important facts for the practical use of this process in

plants transformation. Firstly, the tumour formation is a transformation process of plant

cells resulted from transfer and integration of T-DNA and the subsequent expression of

T-DNA genes. Secondly, the T-DNA genes are transcribed only in plant cells and do

not play any role during the transfer process. Thirdly, any foreign DNA placed between

the T-DNA borders can be transferred to plant cell, no matter where it comes from.

These well etablished facts, allowed the construction of the first vector and bacterial

strain systems for plant transformations (Opabode 2002). Agrobacterium-mediated gene

transfer into monocotyledonous plants became possible when reproducible and efficient

methodologies were established on rice, banana, corn, wheat, and sugarcane (Opabode,

2006).

2.8 BINARY VECTORS FOR AGROBACTERIUM MEDIATED

TRANSFORMATION

In binary vectors, the transfer apparatus (i.e. the vir genes) and the T-DNA are located

on separate plasmids. As only the border sequences are needed to define the T-DNA

region and the vir region is absent, binary vectors are relatively small (and therefore

easily manipulated) (Slater et al., 2003).

2.8.1 pCAMBIA Vector

The pCAMBIA vector backbone is derived from the pPZP vectors (Hajdukiewicz et al.,

1994). pCAMBIA vectors offer several important and beneficial characteristics.

22

They can be rapidly multiplied in Escherichia coli, therefore their high copy number

produce high DNA yields. For high stability of vector in Agrobacterium pVS1 replicon

was designed in pCAMBIA. They are small in size. Depending on the type of plasmid

pCAMBIA they are usually 7-12kb. In these vectors restriction sites have been designed

for modular plasmid modifications and there are small but adequate poly-linkers for

introducing the DNA of interest. For easy bacterial selection, kanamycin or

choloramphenicol resistant genes have been designed in the backbone of these vectors.

The selection of transformed plants with the pCAMBIA vectors can be carried out with

hygromycin B or kanamycin antibiotics as their resistant genes have been inserted in the

pCAMBIA vector backbone. The presence of gusA reporte genes in the pCAMBIA

vector makes the preliminary assessment of construct translation simple as the inserted

genes in pCAMBIA vector can be fused to the gusA reporter genes. (pCambia vectors,

2010).

23

CHAPTER 3: MATERIALS AND METHODS

3.1 MATERIALS

In this research, a plasmid construct that contained an anti-toxoplasmosis scFv gene

(courtesy of YasminLab) (TOXO), a Protease Inhibitor gene (PI) and a gusA gene was

designed and developed as the plasmid pCTOXO-PI. The diagram of the plasmid DNA

construct is shown in Figure 3.1.

.

Figure 3.1: pCTOXO-PI construct

Three scFv genes (scFv3, scFv9 and scFv15) were used in the development of the new

plasmids. The scFv gene was introduced into the phage display vector pCANTAB5E.

The phage display vector pCANTAB5E carrying the scFv gene and the DNA construct

pCTOXO-KDEL were developed in an earlier study conducted by Prof.Dr. Rofina

Yasmin Othman’s laboratory (YasminLab) in University Malaya.

24

Three protease inhibitor genes (PIs) commercially synthesized, including Bowman-

Birk Trypsin Inhibitor (BBTI), Cathepcin D Inhibitor (CDI) and Oryzacystatin Protease

Inhibitor (OCPI). The Three PI genes were synthesized and cloned into pUC57 vector

by the Biosyntech Company. A BamHI sequence was added at the 5’ end of all three PI

genes. Additionally a KDEL gene and Bgl II sequence was added at the 3’ end of the PI

genes.

The plasmid pCAMBIA 1301 was used as the cloning vector for the development of

three desired DNA constructs of pCTOXO-PI. The desired plasmid constructs to be

produced in this research were designated as pCTOXO-BBTI, pCTOXO-CDI and

pCTOXO-OCPI.

The Escherichia coli strain JM109 was obtained from the laboratory to conduct the

bacteria transformation of desired constructs in this research.

The Agrobacterium tumefacien strain LBA4404 was used for the Agrobacterium

mediated transformation of the constructs into plant cells.

The general overview of experiments conducted in this research is shown in Figure 3.2.

25

Figure 3.2: General Overview of experiments conducted in this research.

3.1.1 Preparation of plasmid pCANTAB5E and pCAMBIA 1301

3.1.1.1 Growing Escherichia Coli cells

Fifty µl of frozen Escherichia coli bacterial cells harboring pCANTAB5E carrying

scFv genes were grown in 10ml LB broth supplemented with 100µg/ml ampicillin, at

37˚C for overnight (12-14 hours). Three replicates were prepared for each scFv gene.

Fifty µl of frozen Escherichia coli cells harboring pCAMBIA 1301 were also grown in

10ml LB broth supplemented with 100µg/ml kanamycin at 37˚C for overnight, shaking

at 220 rpm. Four replicates were prepared.

26

3.1.1.2 Plasmid Extraction

The overnight cultures of Escherichia coli cells carrying either pCANTAB5E or

pCAMBIA 1301 were centrifuged at 6000 rpm for 15 minutes and the supernatant was

discarded. Two hundred µl of ice-cold solution I [50 mM glucose, 10 mM EDTA and 25

mM Tris-Cl (pH8)] was added into the pellets in each falcon tube. After resuspending

the pellets in solution I they were transferred into 1.5ml microcentrifuge tubes and 200

µl of freshly made solution II [0.2 M NaOH, 1% SDS] was added into them and

incubated at room temperature for 4 minutes. Next step is to add 200µl of ice-cold

solution III [3M Potassium Acetate and 1% Acetic Acid] into the mixture and incubate

on ice for 15 minutes. After centrifuging the mixture at 13,000 rpm for 10 minutes the

supernatant were transferred into new 1.5ml microcentrifuge tubes and 1µl of RNAse A

was added into each sample and incubated in water bath at 37˚C for 2 hours.

The next step was performing phenol-chloroform extraction method to isolate DNA

content from aqueous solution. First , 600 µl of phenol was added and after centrifuging

at 13,000 rpm for 3 minutes the aqueous layer on top was transferred into new tubes.

Second, 600 µl of chloroform was added into the each collected sample and they were

spin down at 13,000 rpm for 3 minutes. Finally, about 400 µl of the aqueous phase was

collected from each sample tube by pipetting and 40µl of 3M sodium acetate and 1000µl

of absolute ethanol were added in to each tube of samples, and then incubated in ice for

20 minutes. After that they were centrifuged at 13,000 rpm for 15 minutes. The

supernatant was completely discarded and 1ml of 70% ETOH was added. After

centrifuging at 13,000 rpm for 5 minutes the supernatant was discarded and the pellets

were dried in a dryer centrifuge machine for 5 minutes. Finally, 50µl of sterile distilled

27

water (sdH2O) was added into each microcentrifuge tubes to resuspend the pellet of

plasmids pCANTAB5E and pCAMBIA 1301.

3.1.2 Preparation of plasmid pUC-PI

3.1.2.1 Preparation of competent cells

One hundred µl of JM109 culture was grown in 10ml fresh LB broth at 37˚C for

overnight in water bath shaker, shaking at 220rpm to prepare Escherichia coli

competent cells for the transformation of synthesized plasmid construct of pUC57

carrying PI genes (three pUC-PIs: pUC-BBTI, pUC-CDI, pUC-OCPI). After 12-14

hours of incubation ,500µl of fresh cells from overnight culture was transferred into

10ml fresh LB broth and was shaken for another 1-2 hours at 37˚C to achieve the

optical density 0.5 at the wavelength of 600 nm.

After reaching the desired optical density (0.5) the culture was transferred into 15ml

Falcon tube in sterile condition and then incubated on ice for 30 minutes. The

refrigerated centrifuge machine was adjusted to 4˚C and the cultures were centrifuged at

3000rpm for 5 minutes at 4˚C. The supernatant was removed and 5ml of RF1 solution

[100mM KCl, 50Mm MnCl2,30mM Potassium acetate, 10mM CaCl2 and 15%

Glycerol (pH5.8)] was added into the pellets and incubated on ice for 20 minutes before

centrifuging at 3000 rpm for 5 minutes, 4˚C. After centrifugation the supernatant was

removed and appropriate volume of RF2 solution [10mM MOPS, 10mM KCl, 75mM

CaCl2, and 15% Glycerol (pH6.8)] (depending on the pellet) was added into the cells.

The pellets were dissolved in RF2 solution within 20 minutes by tapping the Falcon

tube while keeping on ice until the pellets were totally dissolved. Finally, 100µl of

28

competent cells were aliquot to 1.5ml microcentrifuge tubes and frozen in liquid

nitrogen prior to storing at -20˚C.

3.1.2.2 Transformation, analysis and extraction of three plasmid DNA, pUC-PI,

in E.coli cells

Three tubes of 100µl frozen JM109 bacteria culture which were prepared as described

in Section 3.1.2.1 were thawed in ice for 30 minutes and 3µl of each plasmid DNA,

pUC-PI, carrying BBTI, CDI and OCPI was added into them separately to insert the

plasmid DNA. The tubes were then heat shocked in a water bath at 42˚C for 45 seconds.

The tubes were then rapidly transferred to an ice bath for 5 minutes. Then, 900µl of LB

broth was added to them and incubated for 2 hours at 37˚C in a spin shaker.

Subsequently, 100µl culture from each tube was spread on LB agar plates supplemented

with 100µg/ml ampicillin and incubated at 37˚C for overnight.

Colony selection for the transformed bacteria with pUC-PI plasmid constructs was

carried out after 12 to 16 hours incubation. This procedure was carried out under sterile

conditions. The single bacteria colonies formed in the plates of the transformed cultures

were selected using a sterile toothpick. The single colonies were cultured onto a gridded

DNA library master plate containing LB agar supplemented with 100µg/ml ampicillin.

The same colonies were also re-suspended in 50µl of sterile distilled water in individual

0.5ml PCR tubes. The DNA library master plate was then incubated at 37˚C for

overnight. The colony suspensions in the tubes of distilled water were boiled at 99˚C for

10 minutes before performing colony confirmation PCR.

29

After performing colony PCR as will be described in Section 3.2.1 the positive

transformed colonies that showed the desired band in gel electrophoresis of colony PCR

products were chosen from library plate prepared as described in Section 3.1.2.2 and

grown in 10ml LB broth supplemented with 100µg/ml ampicillin. After 12 to 14 hours

incubation at 37˚C in a water bath shaker the culture of positive colonies were harvested

by centrifuging at 6000 rpm for 15 minutes. Then, the plasmids were extracted using the

same phenol-chloroform method as described earlier in Section 3.1.1.2.

3.1.3 Preparation of plasmid construct pCTOXO-KDEL

The Escherichia coli culture harboring pCTOXO-KDEL plasmid DNA construct

obtained from previous studies was grown in 10ml fresh LB broth supplemented with

50µg/ml kanamycin at 37˚C for overnight. After 14 hours of incubation the plasmid

DNA pCTOXO-KDEL was extracted using the same technique for plasmid preparation

as described in Section 3.1.1.2.

3.2 POLYMERASE CHAIN REACTION

3.2.1 Colony PCR for pUC-PIs

The transformation of plasmid DNA pUC-PI constructs into grown bacteria cells was

verified by performing the colony PCR method. The colony PCR was carried out by

amplifying a segment of the desired PI genes using the Polymerase Chain Reaction

(PCR) under these conditions:

Initial denaturation 1 min 94˚C

30

Denaturation 1 min 94˚C

Annealing 30 sec 59.3˚C

Elongation 1 min 72˚C

Repeat step 2 additional 34 cycles

Final elongation 5 mins 72˚C

Cooling 10 mins 25˚C

The primers that were used for colonies transformed with pUC-BBTI, pUC-CDI and

pUC-OCPI are listed in below:

BBTI-Forward primer 5’-TGC ATC TAG AGG ATC CTA CAG CGT G-3’

BBTI-Reverse primer 5’-GGA TCC AGA TCT CTA GAG CTC ATC T-3’

CDI-Forward primer 5’-TAT CGG ATC CGG ATC CTA CAA TAT A-3’

CDI-Reverse primer 5’-GGC CCA GAT CTC TAG AGC TCA TCT T-3’.

OCPI-Forward primer 5’-TAT CGG ATC CGG ATC CTA CGC ATT C-3’

OCPI-Reverse primer 5’-GAC GGG CCC AGA TCT CTA GAG CTC A-3’

3.2.2 PCR Confirmation for extracted pUC-PI

The extracted pUC57 plasmids carrying BBTI, CDI and OCPI were subjected to PCR

to confirm back the presence of desired PI genes after plasmid preparation. The primers

used and the PCR condition was the same as colony PCR for pUC-PI plasmid DNA

constructs as described in Section 3.2.1.

31

3.2.3 PCR Confirmation for extracted pCTOXO-KDEL

The extracted pCAMBIA 1304 plasmid carrying scFv116-KDEL gene was subjected

to PCR to confirm back the presence of desired gene after plasmid preparation. The

annealing temperature for scFv116-KDEL gene Polymerase Chain Reaction was 61.2˚C

and the primers used were as follows:

scFv-KDEL Forward

primer

5’-GCA TTA AGC TTC GCA ATT CCT TTA GTT

GTT CC-3’

scFv-KDEL Reverse primer 5’-CGC CAT GGT CTG AGA AAG ATG AGC TCT

AGT CCA GAC GTT-3’

3.2.4 PCR for anti-toxoplasmosis scFv gene

The extracted pCANTAB5E plasmid carrying scFv anti-toxoplasmosis gene as

described in Section 3.1.1.2 was subjected to PCR in order to insert the desired NcoI

and BamHI Restriction Endonuclease sites (RE sites) at 5’ and 3’ end respectively,

before performing RE digestion. The annealing temperature was 57.8˚C.

The primers used to amplify three scFv genes are listed in below:

NcoI- scFv Forward primer 5’-CCA TGG TTG GCT GCA GAG ACA GTG-3’

BamHI-scFv Reverse primer 5’-GGA TCC GTT CCT TTC TAT GC-3’

32

After running PCR, the purification of three anti-toxoplasmosis scFv gene PCR

products was carried out using Megaquick- spin kit from iNtRON Company. The

manufacturer instruction was performed. The PCR products of scFv3, scFv9 and scFv15

genes were purified.

3.2.5 PCR for Protease Inhibitor genes (PIs)

Polymerase Chain Reaction was carried out for three PI genes using one set of

primer. All PI genes contained BamHI Restriction Endonuclease site (RE site) at 5’ end

and KDEL-BglII fragment at 3’ end so the purpose of performing PCR was not

inserting the RE sites. The PCR for PI genes was done to get thicker and brighter band

of amplified PI genes that can be easily observed in electrophoresis GEL. The PCR

products then can be excised from the GEL by performing the gel extraction method

which will be described in Section 3.3.4.

The annealing temperature was 55˚C and the primers used were as follows:

PIs-Forward primer 5’-TAA AAC GAC GGC CAG TGA AT-3’

PIs-Reverse primer 5’-ATG ACC ATG ATT ACG CCA AG-3’

Purification of PIs PCR products was carried out using Megaquick- spin kit from

iNtRON Company. The manufacturer instruction was performed and BBTI, CDI and

OCPI PCR products were purified.

33

3.3 RESTRICTION ENDONUCLEASE DIGESTION

3.3.1 Double RE digestion for anti-toxoplasmosis scFv gene

Double RE digestion for scFv genes was carried out with NcoI and BamHI restriction

endonuclease enzymes (Conc. 5u/µl) from Fermentas Company. Two µl of each enzyme

was added to 5µl of pure PCR product of scFv gene prepared earlier as described in

Section 3.2.4 in the presence of 5µl of 10X Tango Buffer. The recommended buffer for

double RE digestion with these two enzymes was 1X Tango Buffer according to

Fermentas website. Then, 36µl of sterile distilled water was added into each tube to

provide total volume of 50µl RE digestion solution. The tubes were incubated at 37˚C in

a water bath for overnight.

3.3.2 Double RE digestion for Protease Inhibitor genes

For double RE digestion of PI genes 2µl of each BamHI and BglII enzyme (Conc.

5u/µl) from Fermentas company was added to 5µl of pure PCR products of PI genes

prepared earlier as described in Section 3.2.5 in the presence of 10µl of 10X Tango

Buffer. The recommended buffer for double RE digestion with BamHI and BglII

enzymes was 2X Tango Buffer according to Fermentas website. Then, 31µl of sterile

distilled water was added into each tube to provide total volume of 50µl RE digestion

solution. The tubes were incubated at 37˚C in a water bath for overnight.

3.3.3 Double RE digestion for cloning vector pCAMBIA 1301

In order to provide the desired cloning site for the insertion of scFv-PI fragment to

pCAMBIA 1301 backbone, 2µl of each NcoI and BglII RE enzymes (Conc. 5u/µl) from

Fermentas company was added to 5µl of prepared pCAMBIA 1301 plasmid as

34

described in Section 3.1.1 in the presence of 10µl of 10X Tango Buffer. The

recommended buffer was 2X Tango Buffer according to Fermentas website. Then, 31µl

of sterile distilled water was added into each tube to provide 50µl of total solution.

Finally, the tubes were incubated at 37˚C in a water bath for overnight.

3.3.4 GEL Extraction

The digested products of scFv genes, Protease Inhibitor genes and pCAMBIA 1301

vector were loaded in a 1% w/v agarose gel with 1X TBE buffer and run at 90 volts for

1 hour. After achieving the complete separation of digested bands, they were visualized

under long wavelength UV lamp in dark room and the desired fragments of DNA were

excised using a clean scalpel.

The desired DNA fragment was purified from the excised gel using an Agarose Gel

DNA Purification Megaquick spin kit from iNtRON Company. The manufacturer

instruction was performed and RE digested scFv genes, PI genes and pCAMBIA 1301

were purified.

3.4 CONSTRUCTION OF PCAMBIA EXPRESSION VECTORS

3.4.1 pCTOXO-BBTI construct

T4 DNA ligase enzyme was applied for the ligation of purified RE digested scFv

gene, BBTI gene and pCAMBIA 1301 plasmid as the cloning vector. Total solution of

10µl ligation mixture was prepared by mixing 4µl of digested pCAMBIA 1301, 2µl of

digested scFv gene and 2µl of digested BBTI gene as described in Section 3.3,

supplemented with 1µl of 10X ligation buffer and 1 µl T4 DNA ligase enzyme.

35

Then, the ligation mixture of plasmid construct pCTOXO-BBTI was transferred into

Escherichia coli JM109 cells. For this procedure one tube of frozen JM109 competent

cell was thawed in ice for 30 minutes and 3µl of pCTOXO-BBTI ligation mixture was

added into Escherichia coli JM109 cells to insert the plasmid DNA carrying the desired

scFv-BBTI genes. The tube was then heat shocked in a water bath at 42˚C for 45

seconds. It was then rapidly transferred to an ice bath for 5 minutes. Then, 900µl of

fresh LB broth was added to it and incubated for 2 hours at 37˚C in a spin shaker. After

incubation, the mixture was centrifuged at 1000 rpm for 10 minutes and 800µl of

supernatant was removed. The pellet was then resuspended in the remaining culture.

Subsequently, 100µl of the culture was spread on LB agar plate supplemented with

100µg/ml kanamycin and incubated at 37˚C for overnight. Figure 3.3 shows the

expected pCTOXO-BBTI construct.

36

Figure 3.3: pCTOXO-BBTI construct

3.4.2 pCTOXO-CDI construct

The procedure for the construction of the pCTOXO-CDI construct was the same as

construction of the pCTOXO-BBTI as described in Section 3.4.1 except for one

component which was the protease inhibitor gene. Therefore, the RE digested BBTI

was replaced with RE digested CDI in the ligation mixture. The expected pCTOXO-

CDI construct is shown in Figure 3.4.

37

Figure 3.4: pCTOXO-CDI construct

3.4.3 pCTOXO-OCPI construct

The procedure for the construction of the pCTOXO-OCPI construct was the same as

construction of two previous constructs as described in Sections 3.4.1 and 3.4.2 except

for the protease inhibitor gene. So the RE digested BBTI /CDI was replaced with RE

digested OCPI in the ligation mixture. The expected pCTOXO-OCPI construct is shown

in Figure 3.5.

38

Figure 3.5: pCTOXO-OCPI construct

3.5 COLONY PCR FOR PCTOXO-PIS CONSTRUCTS

Colony selection for the transformed JM109 bacteria with three pCTOXO-PIs as

described in section 3.4 was carried out after 12 to 16 hours incubation. This procedure

was carried out under sterile condition. The single colonies were cultured onto a gridded

DNA library master plate containing LB agar supplemented with 100µg/ml kanamycin.

The same colonies were also resuspended in 50µl of sterile distilled water in individual

0.5ml PCR tubes. The DNA library master plate was then incubated at 37˚C for

overnight. The colony suspensions in the tubes of distilled water were boiled at 99˚C for

10 minutes before performing colony PCR verification.

39

First, colony PCR was done with scFv primers to select the positive colonies which at

least contained the scFv gene showing correct band size on the electrophoresed gel. The

primers used were scFv-Forward and scFv-Reverse primers; the sequence of the primers

described in Section 3.2.4. After colony PCR with scFv primers, the positive colonies

suspensions which showed the scFv band on electrophoresed gel were chosen to

perform colony PCR using scFv-PI flanking primers to verify the presence of the

desired DNA fragments of scFv-BBTI, scFv-CDI and scFv-OCPI in pCTOXO-PI

constructs of interest in this research. The annealing temperature was 54.9˚C and the

primers used were as follows:

scFv-PI Forward primer 5’-CAT TTG GAG AGA ACA CGG GGG ACT -3’

scFv-PI Reverse primer 5’-GGG TCC TAA CCA AGA AAA TGA A-3’

The procedure of ligation, transformation and colony PCR with scFv-PI primers were

repeated for several times using different purified RE digested scFv3, scFv9, scFv15,

BBTI, CDI, OCPI and pCAMBIA 1301.

3.6 CONSTRUCTION OF A SECOND PCTOXO-CDI-KDEL CONSTRUCT

A second plasmid construct for pCTOXO-CDI was developed in collaboration with a

parallel study (Go Pei See pers. comm.). The cloning vector, scFv gene and restriction

endonuclease RE sites were all changed to obtain the new pCTOXO-CDI-KDEL

construct containing CDI gene fused to KDEL. Besides, new primers were designed to

amplify scFv116 and CDI genes. A new vector pCAMBIA 1304 was used instead of

pCAMBIA 1301 as the cloning vector and it was digested using NcoI and SpeI RE

enzymes. For the construction of this new construct scFv116 was amplified using new

40

primers to insert NcoI RE site at 5’ end and HindIII RE site at 3’ end. Also new PI

flanking primers were designed to insert HindII and SpeI RE sites at 5’ and 3’ end of

CDI protease gene respectively. The pure RE digested scFv116 gene was then ligated to

pure RE digested CDI protease inhibitor gene and cloned into the pCAMBIA 1304

vector by applying T4 DNA Ligase (Figure 3.6). The plasmid for the new construct of

pCTOXO-CDI-KDEL was prepared using the same method as described in Section

3.3.1. The pCTOXO-CDI-KDEL construct is shown in Figure 3.7.

pCAMBIA 1304 NcoI scFv116 HindIII CDI (with KDEL) SpeI pCAMBIA 1304

Figure 3.6: Ligation product of pCTOXO- CDI-KDEL

Figure 3.7: pCTOXO-CDI-KDEL construct

41

3.7 TRANSFORMATION OF CONSTRUCTS INTO A. TUMEFACIENS

Agrobacterium- mediated transformation method was used to transform the desired

gene into the plant cells.

3.7.1 Preparation of Agrobacterium competent cell

The Agrobacterum tumefaciens strain LBA4404 was streaked onto a fresh solid LB

agar plate supplemented with 50µg/ml rifampicin and was incubated at 28ºC for 2 to 3

days in dark. The rifampicin antibiotic was filter sterilized prior to addition. After 3

days, a single colony of the Agrobacterium was picked from solid LB agar plate with a

sterile toothpick for the inoculation and transferred into 3 ml liquid LB broth medium

supplemented with 50µg/ml rifampicin and incubated in water bath shaker at 28˚C, 200

rpm for overnight (16 to 18 hours). One ml of the overnight Agrobacterium culture was

then subcultured into 25ml of LB broth in 50ml Falcon tube. The culture was incubated

at 28ºC in a water bath shaker (200rpm). Every hour, 100µl of the culture was taken for

optical density (O.D.) reading at absorption λ=600nm. The optical density reading was

taken until it reached OD600~ 0.45 to 0.5.

After achieving the OD600nm~0.5, the Agrobacterium culture was incubated on ice for

20 minutes. The refrigerated centrifuge machine was set at 4˚C. Then, the

Agrobacterium culture was centrifuged at 5,000 rpm for 10 minutes at 4˚C. The

supernatant was discarded then the tube was put back on ice . Appropriate volume of

LB broth was added to the tube to resuspend the pellets of Agrobacterium. Then, 100µl

of the prepared Agrobacterium competent cells was aliquot to sterile 1.5ml

42

microcentrifuge tubes. The liquid nitrogen was applied on the tubes prior to storing the

LBA4404 competent cells in -20˚C. All steps were done in sterile condition.

3.7.2 Agrobacterium heat shock transformation

Two tubes of 100µl frozen Agrobacterium strain LBA4404 cells were thawed on ice

and 5µg of each plasmid construct, pCTOXO-CDI and pCTOXO-KDEL carrying the

scFv116-CDI and scFv116-KDEL genes respectively (described in Sections 3.1.3 and

3.6) was added into each tube of LBA4404 competent cells to transform the extracted

plasmid constructs into Agrobacterium. After add in the plasmids the tubes were

directly chilled on ice for 30 minutes. Then, they were frozen in liquid nitrogen for 30

seconds to 1 minute. Heat shock transformation was carried out by transferring tubes

into 37˚C water bath and incubated for 4 minutes. After that the tubes were transferred

into ice for 1 minute. Then, 900µl of fresh LB broth was added into each tube to grow

the Agrobacterium containing the plasmids, and incubated at 28˚C (room temperature)

on a rotary shaker (200rpm) and placed in the dark for 2 to 4 hours. Finally, 100µl of

each Agrobacterium culture carrying pCTOXO-CDI construct and pCTOXO-KDEL

construct was spread on 2 LB Agar plates supplemented with 100µg/ml kanamycin and

50µg/ml rifampicin. The Agar plates were incubated in dark at 28˚C for 2 to 3 days until

colonies appeared.

3.7.3 Colony PCR for transformed Agrobacterium with plasmid DNA constructs

Colony selection for the transformed Agrobacterium with plasmid constructs was

carried out after 3 days of incubation. This procedure was carried out under sterile

conditions. The single colonies were cultured onto a gridded DNA library master plate

43

containing LB agar supplemented with 100µg/ml kanamycin and 50µg/ml rifampicin.

The same colonies were also resuspended in 50µl of sterile distilled water in individual

0.5ml PCR tubes. The DNA library master plate was then incubated at 28˚C in dark for

overnight. The colony suspensions in the tubes of distilled water were boiled at 99˚C for

10 minutes.

Two µl of each boiled colony suspension was used as the DNA template to perform the

colony PCR for transformed Agrobacterium LBA4404 with two constructs to verify the

existence of desired scFv116-CDI and scFv116-KDEL genes in Agrobacterium.

3.7.3.1 Colony PCR verification for transformed Agrobacterium with pCTOXO-

CDI

Colony PCR was carried out for Agrobacterium containing pCTOXO-CDI construct

.The annealing temperature for the PCR was 58.3˚C. The PCR products were

electrophoresed in a 1% w/v agarose gel with 1X TBE buffer and run at 120 volts for 25

minutes (Figure 4.18).

3.7.3.2 Colony PCR verification for transformed Agrobacterium with pCTOXO-

KDEL

The primers which were used for the verification of Agrobacterium transformation

with pCTOXO-KDEL construct were as follows:

Flanking scFv anti-TOXO Forward 5’-TTG GAG AGA ACA CGG GGG ACT C-3’

44

primer

Flanking scFv anti-TOXO Reverse

primer

5’-TCA CCT TCA CCC TCT CCA CT-3’

The annealing temperature for the PCR was 64.3˚C. Then the PCR products were

electrophoresed in a 1% w/v agarose gel with 1X TBE buffer and run at 120 volts for 25

minutes (Figure 4.19).

3.8 PLANTS CELL SUSPENSION

3.8.1 Tobacco cell suspension media preparation

For the subculturing of tobacco cultivar Nicotiana tabacum cell suspension M2 media

was prepared. The concentration of M2 media components were as follows:

Murashige and Skoog medium including vitamins (MS powder) (4.4g/l), Sucrose

(25g/l), 2, 4-dichlorophenoxyacetic acid (2,4-D) (0.5mg/l) and Kinetin (0.2mg/l). Then,

one litter of sterile distilled water was added to dissolve the ingredients. The pH of

media was adjusted to 5.8 by adding very small amount of 1Molar NaOH solution to the

medium. The M2 media was autoclaved at 121˚C for 15 minutes. After autoclaving the

medium was stored in 4˚C.

45

3.8.2 Subculturing of Tobacco cell suspension

The subculture work of tobacco cell suspension was carried out every 7-14 days

depending on the growth of tobacco cells. The procedure of sub-culturing was done in

sterile condition. The cells present in each conical flask were divided into two flasks to

produce enough tobacco cell suspension for the research. First, 40ml of the used media

was removed by pipetting and then 5 to 10ml of the tobacco cell suspension was

transferred into two autoclaved 250ml conical flasks. Then, 50ml of the prepared

sterilized M2 media was added in each flask. The flasks of plant cells were incubated in

a shaker at 80 rpm, 25˚C. All procedures were carried out in sterile conditions.

The subculture work was routinely repeated for a period of 30 days. After this period of

time enough tobacco cell suspension were established that were needed to perform the

next step of the experiment which was Agrobacterium transformation of plant cells.

This amount of cells was needed for the performance of Quantitative GUS fluorometric

assay.

3.9 PREPARATION OF PLANT CELL SUSPENSIONS

For Agrobacterium-mediated transformation of pCTOXO-CDI and pCTOXO-KDEL

into tobacco cells the plants cell suspension were prepared. For each transformation the

cells of 3 conical flasks were mixed by removing the excessive M2 media first and

transferring into a Petri dish on laminar flow. Then, 5ml of plant cell suspension was

transferred into a 50ml Falcon tube by pipetting. Twenty five ml of sterilized M2 media

was subsequently added into them. The ratio volume of cell suspension to M2 media

was adjusted to 1:5. Then, 5ml plant cell suspension was resuspended into 25ml M2

media by the use of pipetting. Then, 1000µl of the mixture was aliquot into 4

46

disposable, polystyrene bottles. Finally, 8000µl of M2 media was added into the cells.

This step was done one day earlier of infection .So each bottle contained 9ml of plant

cell suspension.

For each colony transformation this step was repeated. So to transform each colony (A1,

A2) of Agrobacterium carrying the pCTOXO-CDI construct and (B1, B2) carrying the

pCTOXO-KDEL construct, 4 replicates of tobacco cell suspension were prepared in

disposable, polystyrene bottles. Besides, 4 replicates of tobacco cell suspension were

also prepared to perform the control experiment.

3.10 PREPARATION OF AGROBACTERIUM CULTURE

Two positive Agrobacterium colonies of each construct were grown from the library

plate into 5ml LB broth supplemented with 100µg/ml kanamycin and 50µg/ml

rifampicin to transform the construct of desired genes into plant cells. The pCTOXO-

CDI containing colonies were named as A1 and A2 and the pCTOXO-KDEL

containing colonies were named as B1 and B2.Then they were incubated at 28˚C in a

water bath shaker at 200 rpm for 16 to 18 hours. After 18 hours of incubation, 3ml of

each overnight culture was subcultured into 30ml LB broth in a 50ml Falcon tube. The

tubes were then incubated at 28˚C in a shaker at 200 rpm. Every hour, 100µl of the

culture was taken for optical density (OD) reading at absorption λ=600nm. The optical

density reading was taken until the OD600~ 0.45 to 0.5. After achieving the

OD600nm~0.5 the refrigerated centrifuge machine was set at 4˚C. Then, the

Agrobacterium culture was centrifuged at 5,000 rpm for 10 minutes at 4˚C. The

supernatant was discarded. Then the pellet of Agrobacterium was resuspended in 3ml

fresh M2 media. All the procedures were carried out in sterile conditions.

47

3.11 AGROBACTERIUM-MEDIATED TRANSFORMATION OF PLANT

CELLS

One ml of the prepared Agrobacterium culture in fresh M2 media as described in

Section 3.10 was added into the 9ml of plant cell suspension which was prepared earlier

as described in section 3.9. Four replicates were prepared for each colony. High

efficiency transformation of Agrobacterium into plant cells was promoted using

acetosyringone with the concentration of 100µM. Then, the infected plant cells were

incubated in dark by covering the bottles with aluminum foil to prevent the exposure of

light. The bottles were incubated at 25˚C for 30 minutes on a shaker shaking at 250

rpm. After 30 minutes all the liquid was removed and only cell suspension was

remained in the bottles. The plant cells were washed by sdH2O to remove the excessive

Agrobacterium in the bottles. Finally, the sdH2O was removed by pipetting.

3.11.1 Co-cultivation procedure

The co-cultivation of Agrobcterium and plant cells was carried out by adding 10ml of

MSO media in the bottles. The cell suspension was then incubated in dark at 80 rpm for

4 days. The MSO media (MS basal media) components for co-cultivation work were as

follows:

Murashige and Skoog medium, including vitamins (MS powder) (4.4g/l) and Sucrose

(20g/l). Then, one litter of sterile distilled water was added to dissolve the ingredients.

The pH of media was adjusted to 5.8 for tobacco cells by adding very small amount of

1Molar NaOH solution to the medium. The MSO media was autoclaved at 121˚C for 15

minutes. After autoclaving the medium was stored in 4˚C.

48

3.11.2 Killing Agrobacterium

After 4 days of co-cultivation, the used MSO media was replaced with 10ml fresh

sterilized MSO media supplemented with 50µg/ml cefotaxime on laminar flow. The

plant cell suspension was incubated in dark, shaking at 80 rpm for another 2 days to kill

the Agrobacterium. After killing the Agrobacterium using cefotaxime antibiotic, the

MSO media was removed and 10ml of fresh MSO media was added into each sample.

The plant cells were incubated in dark at 80rpm for another 7 days to give time to the

plant cells for growth.

3.12 ASSAY OF ß-GLUCURONIDASE (GUS) QUANTITATIVE ACTIVITY

BY FLUOROMETRY

The gusA gene was isolated from Escherichia coli (Jefferson et al., 1987) and a

number of gene cassettes are available (e.g. from Clontech) which allow the insertion of

promoter regions adjacent to the gusA gene generating either transcriptional or

translational gene fusions. ß-glucuronidase can be both assayed fluorometrically (using

4-methylumbelliferone glucuronide, MUG, as substrate) and localized in situ

histochemically (using 5-bromo-4-chloro-3-indolyl glucuronide, X-gluc, as substrate).

This research involved quantifying enzyme activity units that normally expressed in

pmol 4-MU released per mg of protein per minute.

GUS

MUG (non-fluorescent) → glucuronic acid+ 4-MU (fluorescent)

49

3.12.1 Preparation of sample, extraction and fluorescence 4-MU quantification

First, the media of transformed tobacco cells was removed by pipetting and only cells

were remained in the bottles. Three replicates for tobacco cells transformed with each

colony A1 and A2 containing pCTOXO-CDI=KDEL construct and three replicates for

each colony B1 and B2 containing pCTOXO-KDEL construct were prepared. The

untransformed tobacco cells of 3 bottles were also harvested to run the control

experiment without Agrobacterium transformation. The harvested tobacco cells were

then frozen by using liquid nitrogen so the cells were made fragile and grinded easily

with mortar and pestle. The GUS Extraction Buffer (GEB) [50mM NaHPO4, pH 7;

10mM 2-mercaptoethanol; 10mM Na2EDTA; 0.1% sodium lauryl sarcosine; 0.1%

Triton X-100] was prepared to extract the protein from the ground tobacco cells

(Jefferson et al., 1987).

The tobacco cells were lysed using GEB. Total protein was extracted by adding 0.5g of

ground tobacco cells to 250µl of GUS extraction buffer. The mixtures were thoroughly

vortex and incubated on ice for 10 minutes. Then they were centrifuged at 12000 rpm,

for 5 minutes at 4ºC. The supernatant was collected from different 1.5ml

microcentrifuge tubes separately and transferred into new tubes. Then, 100µl

supernatant was added to 700 µl GUS extraction buffer for each sample and the

remainder was saved on ice for performing Bradford protein assay. The supernatants

were incubated for 5 minutes at 37ºC. Then, 200 µl pre-warmed GUS assay buffer

(GAB) [2mM 4-Methyl umbelliferyl β-D glucuronide (MUG) in GUS Extraction

Buffer] was added to the mixture. Then the reaction mixture was vortex and incubated

at 37ºC for 90 minutes. After incubation for 90 minutes, the reaction was terminated.

One hundred µl of the reaction mixture was added to 900 µl of 0.2 M carbonate stop

buffer. The addition of Na2CO3 serves the dual purposes of stopping the enzyme

50

reaction and developing the fluorescence of 4-MU, which is about seven times as

intense at alkaline pH. Finally, the unknown sample fluorescence was measured by

using a spectrofluorometer with the emission at 455 nm and excitation at 365 nm and

calculated into GUS specific activity by using 4-methyl-umbelliferone (4-MU) as

standard. Three replicate assays were carried out for each Agrobacterium colony

transformed into tobacco cells.

3.12.1.1 Standard dilution of 4-Methylumbelliferone and relative Fluorescent

Intensity

Standard Dilution of 4-Methylumbelliferone (4-MU) was prepared and Relative

Fluorescent Intensity of different 4-MU dilutions was measured to obtain the equation

of the curve (4-MU vs. Fluorescent Intensity) (Figure 4.22). The equation was needed to

calculate the amount of 4-MU (pmol) in unknown samples.

One mM 4-MU stock solution was prepared by dissolving 19.8mg of 4-

methylumbelliferone (sodium salt), MW=198.20 in 100ml deionised distilled water.

Then 1µM 4-MU stock solution was prepared from the 1mM 4-MU stock solution and

stored at 4˚ C away from light. Subsequently, 4-MU standard measurements for series

of concentrations as shown in table 3.1 were prepared. The Fluorescent Intensity of each

sample was recorded (Table 4.4) using a fluorometer and the standard dilution curve for

4-MU was plotted (Figure 4.22). From the obtained equation of 4-MU standard curve

the enzyme activity of unknown samples was calculated which expressed in pmol 4-MU

released per mg protein per minute (Table 4.8).

51

0.2M Carbonate buffer (ml) 1µM 4-MU stock (µl) Sample conc.(X) (nM)

2.0 0 0

1.9 100 50

1.8 200 100

1.7 300 150

1.6 400 200

Table 3.1: 4-MU Standard Dilutions

3.12.2 Bradford protein determination

Quick Start Bradford Protein Assay Kit from BIORAD was used to perform the

Bradford assay. The Quick Start Bradford protein assay is a simple and accurate

procedure for determining the concentration of protein in solution. The Bradford assay

is a protein determination method that involves the binding of Coomassie Brilliant Blue

G-250 dye to proteins (Bradford, 1976). The dye exists in three forms: cationic (red),

neutral (green), and anionic (blue) (Compton et al., 1985). Under acidic conditions, the

dye is predominantly in the doubly protonated red cationic form (Amax=470nm).

However, when the dye binds to protein, it is converted to a stable unprotonated blue

form (Amax=595nm). It is this blue protein-dye form that is detected at 595nm in the

assay using a spectrophotometer or microplate reader.

In this research, 20µl of the supernatant of unknown samples which was saved on ice as

described in Section 3.12.1 to perform the Bradford protein assay was transferred into

separate clean 1ml disposable cuvettes and then 1ml pre-warmed 1X Bradford Dye

Reagent (included in the kit )was added to each sample. Then, it was mixed gently by

inversion and incubated at room temperature for 5 to 10 minutes before OD reading.

52

The absorbance was read at 595nm for each sample separately using a

spectrophotometer. The protein concentration (mg/ml) of samples was determined by

using the Bradford protein standard curve (Figure 4.23) as will be described in Section

3.12.2.1.

3.12.2.1 Protein standard curve

In any protein assay, the ideal protein to use as standard is a purified preparation of

the protein being assayed. In the absence of such an absolute reference protein, another

protein must be selected as a relative standard. The best relative standard to use is one

that gives a colour yield similar to that of the protein being assayed. The two most

common protein standards used for protein assays are Bovine Serum Albumin (BSA)

and gamma-globulin. With the Quick Start Bradford protein assay, dye colour

development is significantly greater with BSA than with most other proteins, including

gamma-globulin. Therefore, the BSA standard would be an appropriate standard.

The 1X dye reagent was removed from 4˚C storage and let it warm to ambient

temperature to perform the standard assay. The 1X dye reagent was inverted a few times

before use. The 1mg/ml BSA stock solution included in the Quick Start Bradford

Protein Assay Kit was used to prepare the standard dilutions. For the diluents the same

buffer as in the samples was used. So GEB was applied as the diluents. Nine tubes of

diluted protein standard were prepared to perform the 1ml standard assay in disposable

cuvettes. The kit instruction was performed to prepare the dilutions. The BSA volume in

µl was then converted to BSA concentration in mg as shown in table 3.2.

53

Then, each standard solution was transferred into separate 1m disposable cuvettes

supplemented with 900µl of 1X dye reagent and they were inverted. The standard

solutions were incubated at room temperature for 5 to 10 minutes. The absorbance of

standard samples was measured using a spectrophotometer which was set at 595nm.

The instrument was zeroed using blank sample prior to OD reading. The blank sample

was prepared using water and dye reagent. Triplicate standard measurements were

carried out (Table 4.6).

Table 3.2: Bradford Standard Measurements

To determine the protein concentration of unknown samples, standard curve was drawn

by plotting the OD595nm values (y-axis) versus the BSA protein concentration (x-axis)

Tube BSA volume

(µl)

Diluent(GEB)

Volume (µl)

Bradford dye reagent

(µl)

BSA Protein

conc. (mg)

1 0 100 900 0

2 2.5 97.5 900 0.0025

3 5.0 95 900 0.005

4 7.5 92.5 900 0.0075

5 10.0 90.0 900 0.01

6 15.0 85.0 900 0.015

7 20.0 80.0 900 0.02

8 25.0 75.0 900 0.025

9 30.0 70.0 900 0.03

54

(Figure 4.23). From the obtained equation of the standard curve the protein

concentration of unknown samples was calculated (Table 4.7).

55

CHAPTER 4: RESULTS

4.1 PLASMID PREPARATION

4.1.1 Preparation of plasmid pCANTAB5E and pCAMBIA 1301

The plasmid pCANTAB5E carrying scFv3, scFv9 and scFv15 genes and plasmid

pCAMBIA 1301 as the cloning vector were prepared as described in Section 3.1.1. The

extracted plasmids were electrophoresed in a 1% w/v (weight in gram over volume in

milliliter) agarose gel with 1X TBE buffer and run at 120 volts for 25 minutes. The

results obtained showed the presence of the pCANTAB5E and pCAMBIA 1301

plasmid in the electrophoresed gel (Figure 4.1 and 4.2).

Figure 4.1: Plasmid preparations for pCANTAB5E carrying scFv genes

Lane 1, 2, 3: plasmid sample 1, pCANTAB5E –scFv3

Lane 4: 1Kb DNA ladder

Lane 5, 6, 7: Plasmid sample 2, pCANTAB5E –scFv9

Lane 8, 9, 10: Plasmid sample 3, pCANTAB5E –scFv15

1 2 3 4 5 6 7 8 9 10

0.25-1 Kb

1.5-10 Kb

Plasmids pCANTAB5E

carrying scFv ~ 12 Kb

56

Figure 4.2: Plasmid preparations for pCAMBIA 1301

4.1.2 Preparation of plasmid pUC-PI

4.1.2.1 Colony PCR analysis of transformed plasmid pUC-PI into E.coli cells

The plasmid pUC57 carrying Protease Inhibitor genes BBTI, CDI and OCPI, (pUC-

PIs), were transformed into Escherichia coli JM109 cells as described in Section

3.1.2.2. After colony selection the colony PCR was carried out on the colony

suspensions of the transformed Escherichia coli cells as described in Section 3.2.1. The

PCR products were electrophoresed in a 1% w/v agarose gel with 1X TBE buffer and

run at 120 volts for 25 minutes. The results obtained showed the presence of the PI

Lane 1: 1Kb DNA ladder (Appendix M)

Lane 2, 3, 4, 5: Plasmid sample, pCAMBIA 1301

1 2 3 4 5

0.25-1 Kb

1.5-10 Kb

~ 12 Kb

57

genes as a specific band with the expected size of 472bp for BBTI, 577bp for CDI and

90bp for OCPI gene in the electrophoresed gel (Figure 4.3).

Figure 4.3: Colony PCR for pUC-PIs carrying PI genes transformed into JM109

Escherichia coli competent cells. Expected size of PCR product for each PI gene was

shown in the figure.

4.1.2.2 Plasmid pUC-PIs extraction

Four positive colonies of each pUC-PI constructs transformed into Escherichia coli

cells were grown and the plasmids were extracted as described in Section 3.1.2.2. The

Lane 1: 100 bp DNA ladder (Appendix M)

Lane 2-7: colonies A1-A6, carrying BBTI gene

Lane 8: Negative control

Lane 9: 100 bp DNA ladder

Lane 10-15: colonies B1-B6, carrying CDI gene

Lane 16: Negative control

Lane 17: 100 bp DNA ladder

Lane 18-23: colonies C1-C6, carrying OCPI gene

Lane 24: Negative control

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24

600-2500 bp

100-500 bp

BBTI PCR ~472bp

OCPI PCR~90bp

CDI PCR~577bp

58

extracted plasmids were electrophoresed in a 1% w/v agarose gel with 1X TBE buffer

and run at 120 volts for 25 minutes. The results obtained showed the presence of the

plasmid pUC57 in the electrophoresed gel (Figure 4.4). The plasmid pUC57 was used

as the cloning vector for PI genes by the Biosyntech company for after synthesysing the

PI genes as described in Section 3.1.

Figure 4.4: Plasmid preparation for pUC57 vector carrying PI genes (pUC-PI) from

transformed Escherichia coli.

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

1.5-10 Kb

0.25-1 Kb

Lane 1: 1 Kb DNA ladder

Lane 2-5: plasmid prep for pUC-BBTI

Lane 6: 100 bp DNA ladder

Lane 7-10: plasmid prep for pUC-CDI

Lane 11: 100 bp DNA ladder

Lane 12-15: plasmid prep for pUC-OCPI

1 Kb

pUC carrying BBTI ~ 3 Kb pUC carrying CDI ~ 3 Kb pUC carrying OCPI ~ 3 Kb

3 Kb

59

4.1.2.3 PCR Confirmation for extracted pUC-PI

The extracted pUC57 plasmids carrying BBTI, CDI and OCPI were subjected to PCR

to confirm the presence of desired PI genes after plasmid preparation. The primers used

and the PCR condition was the same as colony PCR for pUC57 plasmid as described in

Section 3.2.1. The PCR products were electrophoresed in a 1% w/v agarose gel with 1X

TBE buffer and run at 120 volts for 25 minutes. The results obtained showed the

presence of the PI genes as a specific band with the expected size of 472bp for BBTI,

577bp for CDI and 90bp for OCPI gene in the electrophoresed gel (Figure 4.5).

Figure 4.5: PCR verification for plasmid pUC57 carrying PI genes after plasmid

preparation. Expected size of PCR products: BBTI= 472bp, CDI= 577bp, OCPI=90bp

Lane 1: 100 bp DNA ladder Lane7-10: PCR for plasmid prep pUC-CDI

Lane 2-5: PCR for plasmid prep pUC-BBTI Lane 11: 100 bp DNA ladder

Lane 6: 100 bp DNA ladder Lane 12-15: PCR for plasmid prep pUC-OCPI

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

~472bp

~90bp

~577bp

600-2500 bp

100-500 bp 500 bp

500 bp

60

4.1.3 Preparation of plasmid construct pCTOXO-KDEL

The plasmid pCAMBIA 1304 carrying scFv116-KDEL genes (pCTOXO-KDEL)

were extracted as described in Section 3.1.3. The extracted plasmids were

electrophoresed in a 1% w/v agarose gel with 1X TBE buffer and run at 120 volts for 25

minutes. The results obtained showed the presence of the pCAMBIA 1304 in the

electrophoresed gel. Expected size of pCTOXO-KDEL plasmid was 12 Kb as shown in

Figure 4.6.

Figure 4.6: Plasmid preparation for pCTOXO-KDEL plasmid construct.

1 2 3 4

0.25-1 Kb

1.5-10 Kb

Lane 1: 1 Kb DNA ladder

Lane 2-4: plasmid prep sample, pCTOXO-KDEL

~ 12 Kb

10 Kb

1 Kb

61

4.1.3.1 PCR Confirmation for extracted pCTOXO-KDEL

The extracted pCAMBIA 1304 plasmid carrying scFv116-KDEL gene was subjected

to PCR as described in Section 3.2.3 to confirm back the presence of desired gene after

plasmid preparation. The PCR product was electrophoresed in a 1% w/v agarose gel

with 1X TBE buffer and run at 120 volts for 25 minutes. The results obtained showed

the presence of the scFv116-KDEL genes as a specific band with the expected size of

900bp in the electrophoresed gel (Figure 4.7). This construct was selected for use in the

subsequent transformation experiments.

Figure 4.7: PCR product for pCTOXO-KDEL using scFv-KDEL primers. Expected

size of scFv116-KDEL: 900bp

1 2 3 4 5

Lane 1: 100 bp DNA ladder

Lane 2-4: PCR for pCTOXO-KDEL

Lane 5: Negative control

~900bp

1000-2500

bp

600-900 bp

100-500 bp

62

4.2 PCR FOR ANTI-TOXOPLASMOSIS SCFV AND PROTEASE

INHIBITOR GENES

The PCR was carried out for scFv3, scFv9 and scFv 15 as described in Section 3.2.4

to insert the desired RE sites. The PI genes were also amplified as described in Section

3.2.5. The PCR products were purified using Megaquick-spin kit. The purified PCR

product of scFv3, scFv9 and scFv15 genes and PI genes, BBTI, CDI and OCPI were

electrophoresed in a 1% w/v agarose gel with 1X TBE buffer and run at 120 volts for 25

minutes. The results obtained showed the presence of the scFv genes as a specific band

with the expected size of 800bp in the electrophoresed gel (Figure 4.8). The BBTI, CDI

and OCPI protease inhibitors were also shown very distinct and clear band of 700bp,

950bp and 800bp respectively (Figure 4.9).

Figure 4.8: Purified PCR product of anti-toxoplasmosis scFv genes using scFv primers.

Expected size: scFv=800bp

Lane 1: 100bp DNA ladder

Lane 2: PCR product for scFv3 gene

Lane 3: PCR product for scFv9 gene

Lane 4: PCR product for scFv15 gene

1 2 3 4

~ 800bp

100-500 bp

600-2500 bp

500bp

63

Figure 4.9: Purified PCR product of PI genes using PI primers.

Expected size of PCR products: BBTI= 700bp, CDI= 950bp, OCPI= 800bp.

4.3 DOUBLE RE DIGESTED GENES

Three scFv genes, PI genes and pCAMBIA 1301 cloning vector were digested using

restriction endonuclease enzymes to produce desired fragmented DNA as described in

Section 3.3. The digested genes were gel extracted and purified as described in Section

3.3.4. After purification process was done each purified DNA fragment was

electrophoresed in a 1% w/v agarose gel with 1X TBE buffer and run at 120 volts for 25

Lane 1: 100 bp DNA ladder

Lane 2: PCR product for BBTI gene

Lane 3: PCR product for CDI gene

Lane 4: PCR product for OCPI gene

1 2 3 4

~700bp ~800bp

~950bp

100-500 bp

600-900 bp

1000-2500bp

64

minutes. The results obtained showed the presence of the pure digested scFv genes, PI

genes and pCAMBIA 1301 plasmid as a specific band in the electrophoresed gel

(Figure 4.10 and 4.11).

Figure 4.10: Purified RE digested scFv genes and pCAMBIA 1301

Expected size of RE digested genes: scFv: 760bp, pCAMBIA 1301:12Kb

1 2 3 4 5 6 7

~ 760bp

~ 12Kb

100-500 bp

600- 2500bp

1Kb

10Kb

Lane 1: 100 bp DNA ladder

Lane 2: RE digested scFv3 gene

Lane 3: RE digested scFv9 gene

Lane 4: RE digested scFv15 gene

Lane 5: 1 Kb DNA ladder

Lane 6-7: RE digested pCAMBIA 1301

65

Figure 4.11: Purified RE digested Protease Inhibitor genes

Expected size of RE digested PI genes: BBTI=591BP, CDI=834bp, OCPI=679bp

4.4 CONSTRUCT ANALYSIS

4.4.1 Analysis of pCTOXO-PIs

4.4.1.1 PCR analysis of transformed E.coli with ligation mixture of pCTOXO-PIs

PCR was carried out on colony suspension of transformed JM109 Escherichia coli

cells with pCTOXO-PIs as described in Section 3.5. First, colony PCR was carried out

with scFv primers to confirm wherever the colonies carried scFv gene. The PCR

products were electrophoresed in a 1% w/v agarose gel with 1X TBE buffer and run at

Lane 1: 100 bp DNA ladder

Lane 2: RE digested BBTI gene

Lane 3: RE digested CDI gene

Lane 4: RE digested OCPI gene

1 2 3 4

100-500 bp

500-2500 bp

~ 591bp ~ 679bp

~ 834bp

66

120 volts for 25 minutes. The results obtained showed the presence of the scFv genes as

a specific band with the expected size of 750bp in the electrophoresed gel (Figure 4.12

and 4.13). The unexpected bands showed as PCR product in GEL electrophoresis result

(Figure 4.12) might be due to primer dimer problem. The scFv primers using in this step

might have strings of complimentary bases that can attach to each other and lead to a

potential by-product in PCR.

Figure 4.12: Colony PCR for pCTOXO-PIs using scFv primers.

scFv15+PIs+pCAMBIA1301

1 2 3 4 5 6 7 8 9 10 11 12 13

A

~750bp

100-500 bp

500-2500 bp

Lane1: 100 bp DNA ladder

Lane 2-4: PCR for pCTOXO-BBTI

Lane 5-9: PCR for pCTOXO-CDI

Lane 10-13: PCR for pCTOXO-OCPI

Unexpected bands

500bp

67

Figure 4.13: Colony PCR for pCTOXO-PIs using scFv primers.

scFv9+BBTI+pCAMBIA1301

The positive colony suspensions that showed the scFv band on electrophoresis gel were

then chosen to perform the colony PCR using scFv-PI flanking primers to confirm the

success of ligation process. Then the PCR products were electrophoresed in a 1% w/v

agarose gel with 1X TBE buffer and run at 120 volts for 25 minutes. The results

obtained did not show the presence of the correct band size for amplified scFv-PI genes.

The expected size of PCR product for ligated scFv-BBTI gene was 1400bp but the gel

picture showed a band with the size of about 850bp (Figure 4.14). For scFv-CDI the

expected size was 1600bp but the obtained result showed a band with the size of 850bp

(Figure 4.15). Also for the scFv-OCPI the expected size of the PCR product was

B

1 2 3 4 5 6

~750bp

100-500 bp

500-2500 bp

Figure 4.13:

Lane1: 100 bp DNA ladder

Lane 2-6: PCR for pCTOXO-BBTI

68

1450bp. However, the result showed a band with 850bp in the electrophoresed gel

(Figure 4.16).

Figure 4.14: Colony PCR for pCTOXO-BBTI using scFv-PI flanking primers.

Expected size of pCTOXO-BBTI: 1400bp

Lane 1: 100 bp DNA ladder

Lane 2-6: PCR for pCTOXO-BBTI

1 2 3 4 5 6

100-500 bp

500-2500 bp

~ 850bp

69

Figure 4.15: Colony PCR for pCTOXO-CDI using scFv-PI flanking primers.

Expected size of pCTOXO-CDI: 1600bp

Figure 4.16: Colony PCR for pCTOXO-OCPI using scFv-PI flanking primers.

Expected size of pCTOXO-OCPI: 1450bp

Lane 1: 1 Kb DNA ladder

Lane 2-9: PCR for pCTOXO-CDI

1 2 3 4 5 6 7 8 9

0.25-1 Kb

1.5-10 Kb

~850bp

Lane 1: 1 Kb DNA ladder

Lane 2-8: PCR for pCTOXO-OCPI

1 2 3 4 5 6 7 8

0.25-1 Kb

1.5-10 Kb

~850bp

70

The procedure of ligation, transformation and colony PCR with scFv-PI primers were

repeated for several times using different purified RE digested scFv3, scFv9, scFv15,

BBTI, CDI, OCPI and pCAMBIA 1301. But the result obtained still did not show the

desired correct band size of PCR product for ligated scFv-PI genes.

4.4.1.2 Gene Sequencing Analysis

The PCR was carried out on two pCTOXO-PIs colony suspensions prepared after

transforming the ligation mixture of pCTOXO-BBTI into JM109 cells. The PCR was

carried out in large scale, 50µl, for scFv-PI genes using scFv-PI flanking primers. The

sequence of primers was described in Section 3.5. The PCR products were purified with

the use of Megaquick-spin kit from iNtRON Company. The purified PCR products were

then sent for sequencing.

The gene sequencing results were analyzed using scFv-PI flanking primer sequences to

search for the desired sequence of scFv-PI gene ligated to the pCAMBIA 1301, but the

obtained result showed only scFv anti-toxoplasmosis gene ligated to the pCAMBIA

1301vector without ligating to Protease Inhibitor gene. (Gene Sequencing results in

Appendix A).

The analysis of forward DNA sequencing results was carried out (Appendix B) by

searching the sequence with the reverse complement sequence of scFv-PI reverse

primer. The reverse complement sequence of scFv-PI reverse primer was as follows:

5’-TTCATTTTCTGTTTTAGGAACCCA - 3’

71

Then, the reverse DNA sequencing result was first reverse complemented to search for

the desired scFv-PI gene sequence using forward scFv-PI primer (Appendix C). The

two obtained forward DNA sequence and reverse DNA sequence was then combined

for searching for the desired scFv-PI sequence ligating to the pCAMBIA 1301vector.

The Appendix B shows the forward DNA sequencing result and Appendix C shows the

reverse DNA sequencing result. The combination of these two sequences was shown in

Appendix D and the scFv sequence and pCAMBIA 1301 sequence were highlighted but

the PI gene sequence was not identified as it was not ligated to the vector.

4.4.2 pCTOXO-CDI-KDEL

As the previous attempts to obtain the recombinant PI constructs did not yield any

suitable clone a new pCTOXO-CDI construct containing the scFv116-CDI gene

developed with a parallel project in YasminLab (Go Pei See pers.comm. as described in

Section 3.6) was used for the transformation experiments. The construct also contained

a KDEL element and hence was labeled pCTOXO-CDI-KDEL.

4.4.2.1 PCR analysis of extracted pCTOXO-CDI from E.coli cells

The extracted pCTOXO-CDI-KDEL plasmid was used to perform the

Agrobacterium-mediated transformation of plant cells in this study. The PCR was

carried out on the new pCTOXO-CDI-KDEL plasmid construct using scFv-PI flanking

primers to confirm the presence of scFv116-CDI gene after plasmid preparation. The

PCR products were electrophoresed in a 1% w/v agarose gel with 1X TBE buffer and

run at 120 volts for 25 minutes (Figure 4.17)

72

Figure 4.17: PCR for ligated plasmid construct pCTOXO-CDI2 using new scFv-PI

primers.Expected size of PCR product: 1.5 Kb

4.5 PCR ANALYSIS OF TRANSFORMED AGROBACTERIUM LBA4404

WITH PLASMID CONSTRUCTS

Colony PCR was carried out for Agrobacterium transformed with pCTOXO-CDI and

pCTOXO-KDEL to verify the presence of desired constructs. The PCR products were

electrophoresed in a 1% w/v agarose gel with 1X TBE buffer and run at 120 volts for 25

minutes. The results obtained showed the presence of the scFv116-CDI gene as a

specific band with the expected size of 1500bp (Figure 4.18) and scFv116-KDEL gene

as a specific band with the expected size of 1100bp in the electrophoresed gel of some

transformed Agrobacterium colonies (Figure 4.19).

1 2 3 4 5

~I.5 Kb

Lane 1: 100 bp DNA ladder

Lane 2-5: PCR for pCTOXO-CDI construct

100-500 bp

600-2500 bp

73

Figure 4.18: Colony PCR for Agrobacterium transformed with pCTOXO-CDI plasmid

construct. Expected size: 1500 bp

Lane 1: 100 bp DNA ladder Lane 6: positive control

Lane 2: transformed colony A1 Lane 7: negative control

Lane 3-4: untransformed colonies

Lane 5: transformed colony A2

1 2 3 4 5 6 7

100-500 bp

600-2500 bp

~ 1500bp

74

Figure 4.19: Colony PCR for Agrobacterium transformed with pCTOXO-KDEL

plasmid construct. Expected size: 1100 bp

4.6 TARGET SYSTEM

4.6.1 Tobacco cell suspension culture

In this research, the target system used was cell suspension of Nicotiana tabacum cv.

BY-2 (cultivar Bright Yellow - 2 of the tobacco plant) for performing the

Lane 1: 100 bp DNA ladder

Lane 2: transformed colony B1

Lane 3: Transformed colony B2

Lane 4: positive control

Lane 5: negative control

1 2 3 4 5

~ 1100bp

100-500 bp

600-2500 bp

75

Agrobacterium-mediated transformation of the two constructs pCTOXO-CDI-KDEL

and pCTOXO-KDEL. The cell suspension was propagated by subculturing as described

in Section 3.8.4. Several flasks of tobacco BY-2 cell suspension were prepared. The

typical tobacco cell suspension is shown in Figure 4.20.

Figure 4.20: Nicotiana tabacum cv. BY-2 cell suspension. Scale bar represents 1 cm.

For each transformed Agrobacterium colony carrying the desired plasmid construct 4

replicates of tobacco cell suspension were prepared. Two colonies for each plasmid

construct were chosen to be transformed into tobacco cells as described in Section 3.9.

The cells were transformed with two plasmid construct pCTOXO-CDI-KDEL and

pCTOXO-KDEL using Agrobacterium-mediated transformation method as described in

Section 3.11. The survival rate of tobacco cell suspension transformed with

Agrobacterium carrying plasmid construct pCTOXO-CDI-KDEL and pCTOXO-KDEL

is summarized in table 4.1.

76

Condition of

cultures

tobacco cell suspension co-

cultivated with pCTOXO-CDI-

KDEL (number of plates used)

tobacco cell suspension co-cultivated

with pCTOXO-KDEL

(number of plates used)

Agrob colony

name

A1 A2 B1 B2

Living plates 4 3 3 4

Contaminated

plates

0 1 1 0

Total plates 4 4 4 4

% survival 100 75 75 100

Table 4.1: Number and percentage of surviving and dead tobacco cell suspension

cultures after 14 days of co-cultivation with Agrobacterium carrying plasmid construct

pCTOXO-CDI-KDEL and pCTOXO-KDEL.

To compare the GUS enzyme activity results obtained by performing the Quantitative

GUS Fluorometric Assay of transformed tobacco cells (A1 vs. B1 and A2 vs. B2), three

living replicate of tobacco cells transformed with each colony were chosen to continue

the experiment to the next step, because the number of samples was required to be equal

for the analysis of data in paired t-test method. Therefore, 1 plate of tobacco cells

transformed with A1 and 1 plate of tobacco cells transformed with B2 were set aside to

have 3 replicates in total from each transformation event.

4.7 QUANTITATIVE GUS FLUOROMETRIC ASSAY

ß-glucuronidase (GUS) is the reporter enzyme of choice for most plant

transformation research. Typically GUS activity in solution is determined with

the fluorogenic substrate 4-methylumbelliferyl ß-D glucuronide (MUG):

77

The reaction product 4-methylumbelliferone (4MU) is maximally fluorescent at high

pH, where the hydroxyl group is ionized. Addition of a basic solution of sodium

carbonate simultaneously stops the assay and adjusts the pH for quantitating the

fluorescent product.

In this research the unknown sample fluorescence (relative fluorescent intensity of 4-

MU) was measured for each sample by using a spectrofluorometer with the emission at

455 nm and excitation at 365 nm as described in Section 3.12.1. The relative fluorescent

intensity of unknown samples is listed in Table 4.2. For the calculation of exact

fluorescent intensity the average amount of control experiments was subtracted from

test samples ‘relative fluorescent intensity amounts to reduce the background effect

which is due to intrinsic effects of plant cells (Table 4.2).

Sample Control Tobacco cells

transformed with

pCTOXO-CDI-KDEL

Tobacco cells

transformed with

pCTOXO-KDEL

Exact Fluorescent Intensity

colony A1 A2 B1 B2 A1 A2 B1 B2

replicate 1 3274 8019 8008 5413 5417 4743 4732 2137 2141

replicate 2 3305 7886 8022 5429 5435 4610 4746 2153 2159

replicate 3 3249 8031 7989 5396 5414 4755 4713 2120 2138

Average 3276 7978.6 8006.3 5412.6 5422 4702.6 4730.3 2136.6 2146

Standard

deviation

28.05352

80.47567

16.56301

16.50253

11.35782

80.47567

16.56301

16.50253

11.35782

Table 4.2: The relative fluorescent intensity of test samples for three replicates of each

colony transformation A1, A2, B1, B2

GUS

MUG (non-fluorescent) → glucuronic acid+ 4-MU (fluorescent)

78

For calculation of 4-MU amount of the test samples in pmol the Standard Dilution of 4-

Methylumbelliferone was carried out as described in Section 3.12.1.1 and the

fluorescent intensity of different dilutions of 4-MU was obtained as listed in Appendix

I.

The measured fluorescent intensity amounts of 4-MU dilutions were plotted versus

different 4-MU concentrations as shown in Appendix J. From the obtained equation of

the curve the Relative Fluorescent Intensity of the test samples was calculated in pmol

as shown in Table 4.3.

Sample Exact Fluorecsent Intensity 4-MU (pmol)

A1 A2 B1 B2 A1 A2 B1 B2

replicate

1

4743 4732 2137 2141 778.79 775.85 352.45 353.11

replicate

2

4610 4746 2153 2159 755.94 778.13 355.06 356.04

replicate

3

4755 4713 2120 2138 779.60 772.75 349.68 352.62

Average 4702.66 4730.33 2136.66 2146 771.44 775.57 352.39 353.92

Standard Deviation (STDEV) 13.43 2.70 2.69 1.84

Table 4.3: Relative Fluorescent Intensity of test samples (pmol)

Comparison of the standard deviation of A1 vs. A2 shows higher STDEV for A1

samples (13.43 > 2.70). However the mean values (average) of A1 and A2 are very

close (Table 4.3). This indicates the 4-MU measurements of A2 samples in three

79

replicates have closer values in comparison with three A1 replicates. Thus the 4-MU

measurements were more accurate in A2 sample replicates.

Comparison of the standard deviation of B1 vs. B2 shows higher STDEV for B1

samples (2.69 > 1.84). However the mean values (average) of B1 and B2 are very close

(Table 4.3). This indicates the 4-MU measurements of B2 samples in three replicates

have closer values in comparison with three B1 replicates. Thus the 4-MU

measurements were more accurate in B2 sample replicates.

4.7.1 Bradford assay

The protein standard assay was carried out as described in Section 3.12.2.1 to obtain

the protein content of the unknown samples. The obtained OD values for different BSA

protein concentrations listed in Appendix K. Then, the protein standard curve was

plotted to obtain the equation needed to determine the protein content of unknown

samples (Appendix L).

The Bradford protein assay was carried out to obtain the amount of soluble protein in

the unknown samples. The absorbance was read at 595nm for each unknown sample

separately using a spectrophotometer as described in Section 3.12.2. Then, the protein

concentration of unknown samples was determined by using the Bradford protein

standard curve (Appendix L). The OD reading values and protein concentration of

samples are listed in table 4.4. Statistical analysis of total soluble protein concentration

in transformed tobacco cells with two plasmid constructs showed significant difference

(Table 4.4). The testing of paired differences between TSP of transformed tobacco cells

80

with pCTOXO-CDI-KDEL and pCTOXO-KDEL were done using paired t-test at P =

0.05.

Sample OD (A595nm) OD (A595nm) Protein conc. mg Protein conc. mg

A1 A2 B1 B2 A1 A2 B1 B2

Replicate 1 0.7310 0.7284 0.6349 0.6382 0.0242 0.0238 0.0104 0.0109

Replicate 2 0.6989 0.7326 0.6412 0.6423 0.0196 0.0244 0.0113 0.0115

Replicate 3 0.7598 0.6983 0.6270 0.6354 0.0337 0.0195 0.0093 0.0105

Average 0.7299 0.7197 0.6343 0.6386 0.0258 0.0225 0.0103 0.0109

Standard Deviation (S.D.) 0.00719 0.00267 0.00100 0.00050

T-TEST protein conc. (mg) A1 vs. B1 0.040846

T-TEST protein conc. (mg) A2 vs. B2 0.006164

Table 4.4: The OD values for test samples and protein conc. of samples

Comparison of the standard deviation of A1 vs. A2 shows higher STDEV for A1

samples (0.00719 > 0.00267). However the mean values (average) of A1 and A2 are

very close (Table 4.4). This indicates the total soluble protein concentration

measurements of A2 samples in three replicates have closer values in comparison with

three A1 replicates. Thus the TSP measurements were more accurate in A2 sample

replicates.

Comparison of the standard deviation of B1 vs. B2 shows higher STDEV for B1

samples (0.00100 > 0.00050). However the mean values (average) of B1 and B2 are

very close (Table 4.4). This indicates the total soluble protein concentration

81

measurements of B2 samples in three replicates have closer values in comparison with

three B1 replicates. Thus the TSP measurements were more accurate in B2 sample

replicates.

The soluble protein concentration mean values (average) of three sample replicates was

plotted in Figure 4-21 and error bars were indicated on the chart.

Figure 4-21: Comparison of TSP of tobacco cells transformed with pCTOXO-CDI (A1

& A2 colonies) and pCTOXO-KDEL (B1 & B2 colonies).

4.8 COMPARISON OF ENZYME ACTIVITY

The gusA gene enzyme activity was quantified and expressed in pmol 4-MU released

per mg of protein per minute according to 4-MU (pmol) and protein conc. (mg) that

were obtained and listed in Tables 4.3 and 4.4 respectively. The 4-MU enzyme activity

was calculated for samples after 90 minutes reaction as described in Section 3.12.1 and

listed in Table 4.5.

0

0.005

0.01

0.015

0.02

0.025

0.03

0.035

mean value of Prot Conc. (mg)

Tobacco cells transformed with

pCTOXO-CDI(A1) pCTOXO-CDI(A2) pCTOXO-KDEL(B1) pCTOXOKDEL(B2)

A1 A2 B1 B2

82

Statistical analysis of GUS activity in transformed tobacco cells with two plasmid

constructs did not show any significant difference (Table 4.5). The testing of paired

differences between GUS activity of transformed tobacco cells with pCTOXO-CDI-

KDEL and pCTOXO-KDEL were done using paired t-test at P = 0.05.

Table 4.5: Comparison of 4-MU enzyme activity between A1 vs. B1 and A2 vs. B2

samples.. 4-MU enzyme activity expressed in pmol/mg. protein/min for tobacco cells

transformed with pCTOXO-CDI-KDEL (A1 and A2 Agrob colonies) and pCTOXO-

KDEL (B1 and B2 Agrobacterium colonies).

Sam

ple

4-MU (pmol) 4-MU

(pmol)

Protein conc.

mg

Protein conc.

mg

4-MU

(pmol)/mg

prot/min

4-MU

(pmol)/mg

prot/min

A1 A2 B1 B2 A1 A2 B1 B2 A1 A2 B1 B2

1 778.79 775.85 352.45 353.11 0.0242 0.0238 0.0104 0.0109 357.571 362.208 376.549 359.949

2 755.94 778.13 355.06 356.04 0.0196 0.0244 0.0113 0.0115 428.537 354.339 349.124 344.000

3 779.60 772.75 349.68 352.62 0.0337 0.0195 0.0093 0.0105 257.039 440.313 417.777 373.143

Av

erag

e

771.44 775.57 352.39 353.92 0.0258 0.0225 0.0103 0.0109 347.71 385.62 381.15 359.03

Standard Deviation 86.172

47.528

34.556

14.593

T-TEST enzyme activity (4-MU: pmol/mg/min) A1 vs. B1 0.339395

T-TEST enzyme activity (4-MU: pmol/mg/min) A2 vs. B2 0.16136

83

The GUS activity mean values (average) expressed in 4-MU/mg protein/min of three

sample replicates was plotted in Figure 4-22 and error bars were indicated on the chart.

Figure 4.22: Comparison of the level of GUS Activity of the transformed tobacco cells

with two different plasmid constructs.

310

320

330

340

350

360

370

380

390

400

A1 A2 B1 B2 Tobacco cells transformed with

pCTOXO-CDI(A1) pCTOXO-CDI(A2) pCTOXO-KDEL(B1) pCTOXO-KDEL(B2)

Mean value of GUS Activity

pmol MU/mg protein/min

84

CHAPTER 5: DISCUSSION

In this research tobacco cell suspension were selected as host systems for testing of

scFv constructs enhanced using protease inhibitor elements. Although no positive

results were obtained using the banana cell suspensions, the scFv anti-toxoplasmosis

gene ligated to Cathepsin D Inhibitor in pCTOXO-CDI-KDEL and also ligated to

KDEL fragment in pCTOXO-KDEL was shown to be putatively transformed and

expressed in tobacco cells as demonstrated by GUS fluorometric Assay.

The gene transfer and expression technology in tobacco cells is well-established. The

high biomass yield is one of the major advantages of tobacco plant as a host system. So

it can be a major source of plant-derived recombinant antibodies. (Twyman et al.,

2007).

The effect of Plant Protease Inhibitor (CDI) on the expression of the heterologous scFv

anti-toxoplasmosis gene was then investigated by comparing the total soluble protein

(TSP) and GUS activity in tobacco cells transformed with two different constructs with

protease inhibitor (pCTOXO-CDI-KDEL) or with the ER-targetting element KDEL(

pCTOXO-KDEL). The measurement of total soluble protein content in the transformed

cells and GUS assay analysis can only assess the preliminary positive effects of CDI

protease inhibitor in recombinant protein production in plant cells, but as these methods

are preliminary assessments for measuring positive transformation events, presence of

higher total soluble protein content (Table 4.4 and Figure 4.21) in cells transformed

with TOXO gene may not necessarily resulted from the effect of the CDI protease

inhibitor.

85

5.1 PLASMID CONSTRUCT DEVELOPMENT

In this research the development of three pCTOXO-PI plasmid constructs expressed

as pCTOXO-BBTI, pCTOXO-CDI and pCTOXO-OCPI was attempted by ligating RE

digested scFv anti-toxoplasmosis and PI genes DNA fragments and cloning into

pCAMBIA 1301vector.

5.1.1 Verification of Ligation

PCR was carried out on ligation mixture using scFv primers and scFv-PI flanking

primers as described in Section 3.5. The PCR products amplified using scFv primers

confirmed the presence of scFv gene ligated to pCAMBIA 1301 vector on gel

electrophoresis (Figure 4.12 and 4.13). However, the results obtained from PCR work

with scFv-PI flanking primers unexpectedly did not show the presence of correct band

size of ligated scFv-PI product in PCR gel electrophoresis (Figure 4.14, 4.15 and 4.16).

The failure of desired plasmid constructs pCTOXO-PI development was also verified

by sequencing analysis as was described in Section 4.4.1.2. The sequencing results

showed the presence of scFv gene ligated to pCAMBIA 1301 vector but the PI gene

sequence was missing in the sequencing result (Appendix D).

5.1.1.1 Failure of plasmid constructs pCTOXO-PI development

The ligation of scFv gene to pCAMBIA 1301 vector was verified by sequencing as

described in Section 4.4.1.2. But no ligation of the PI genes was detected. This failure

could be due to mismatching of compatible sticky ends (Clark, 2005). BamHI and BglII

generate the same overhanging or sticky ends. It means BamHI and BglII generate

86

different-but-compatible ends. BamHI recognizes the sequence 5’-GGATCC-3’ and

cuts after the first 5’ G, which generates the 3’-CTAG-5’ overhang on the bottom

strand. BglII recognizes the sequence 5’-AGATCT-3’ and cuts after the first 5’ A,

which generates a 5’-GATC-3’ overhang on the top strand. If these two pieces are

allowed to anneal, the complementary sequences will hydrogen bond together, allowing

the nicks to be sealed more easily by DNA ligase as shown in Figure 5.1. In this

research the scFv gene had BamHI sticky ends at 5’ end and the vector pCAMBIA 1301

was digested using NcoI and BglII as described in Section 3.3.3. Therefore, the two

BamHI and BglII sticky ends might ligate together despite the fact that BamHI and

BglII generate different ends. Missing (deletion or point mutations) of some parts of

nucleotide sequences in PI genes or pCAMBIA 1301 backbone could be the other

probability that the desired constructs of pCTOXO-PI failed to develop. Because of this

another construct was developed in a parallel study (pCTOXO-CDI-KDEL) and was

used for comparative analysis with the constructed pCTOXO-KDEL.

87

Figure 5.1: Matching of compatible sticky ends

5.2 COMPARISON OF TOTAL SOLUBLE PROTEIN

The Total Soluble Protein (TSP) of Agrobacterium-mediated transformed tobacco

cells with two plasmid constructs, pCTOXO-CDI-KDEL and pCTOXO-KDEL was

measured using Bradford protein assay as described in Section 3.12.2. The paired t-test

(at P=0.05) results of protein concentration (mg) obtained from Table 4.4 indicates a

significant difference (P < 0.05) in TSP of tobacco cells transformed with two different

constructs. The TSP mean values of transformed tobacco cells with two constructs are

illustrated in Figure 4.21 to compare the TSP amounts. The Figure 4.21 indicates that

the TSP of tobacco cells transformed with pCTOXO-CDI-KDEL was higher than

tobacco cells transformed with pCTOXO-KDEL. The CDI gene in this construct was

synthesized with a KDEL fragment at its 3’ end. Hence both plasmid constructs

contained the KDEL fragment but the difference was in pCTOXO-CDI which contained

the Protease Inhibitor gene Cathepsin D Inhibitor. This suggested that the CDI gene had

88

an effect on the TSP in tobacco cells transformed with pCTOXO-CDI. The CDI gene

was suggested to inhibit the endogenous proteases of tobacco cells that degrade the

heterologous protein in plant cells leading to higher TSP measurements in cells carrying

it. These results concurred with a report by Michaud et al., (2005) and Rivard et al.,

(2006). Hence suggesting this strategy would be suitable for enhancing the production

of anti-toxoplasmosis antibodies in plant cells.

5.3 COMPARISON OF ENZYME ACTIVITY

The GUS reporter gene transformation efficiencies were monitored by plasmid

pCAMBIA1304 transformation into cell suspension of tobacco cultivar BY-2 via

Agrobacterium-mediated transformation. The evaluation of the effect of plant protease

inhibitor gene (CDI) on the expression of heterologous gene (scFv anti-toxoplasmosis)

in tobacco cells was based on quantitative fluorometric assay analysis of GUS

expression.

The ß-glucuronidase was visualized as fluorescence blue color in tobacco cell extracts

indicating positive transformation event as described in Section 3.12.1. Since the ß-

glucuronidase was designed as a fusion construct with the gene of interest in the

plasmid construct therefore, observation the presence of ß-glucuronidase could imply

the possible successful expression of scFv-CDI-KDEL and scFv-KDEL genes in the

transformed tobacco cells. Reporter genes like ß-glucuronidase encode for messages

which would clearly indicate the transient or stable expression of the transferred genes

in the transgenic cells (Slater et al., 2003).

In this research, the quantitative fluorometric assays of GUS activity demonstrated the

transcript level of GUS gene in the putative transformed tobacco cells with plasmid

89

pCTOXO-CDI-KDEL and pCTOXO-KDEL. The presence of ß-glucuronidase enzyme

will cleave the fluorogenic substrate 4-methylumbelliferyl β- D glucuronide (MUG) to

produce fluorescent 4- methylumbelliferone (4-MU) (Jefferson et al., 1987). The

Agrobacterium transformed tobacco cells were incubated for two weeks to allow cell

growth and stable gusA gene integration before fluorometric assay was performed.

Figure 4.22 indicates that the transient gene expression level was detected

fluorometrically in tobacco plant cell extracts prepared two weeks after Agrobacterium-

mediated transformation experiment, regardless of the plasmid construct used. In

comparison to the transformed tobacco cells, higher total protein (mg) production level

(TSP) was achieved for pCTOXO-CDI-KDEL transformed cells base on Bradford assay

(Table 4.4 and Figure 4.21). However the statistical analysis (paired t-test) of the GUS

activity (pmol 4-MU/mg protein/min) for the putative transformed tobacco cells

revealed no significant (P > 0.05) differences between pCTOXO-CDI-KDEL and

pCTOXO-KDEL (Table 4.5). This finding was probably due to inadequate sample

replicates for performing paired t-test. Future work should include more replicates to

obtain more statistically accurate results.

These preliminary results suggested that protease inhibitor genes may present a viable

strategy for enhancing levels of production of scFv proteins in plant cells. Further

studies are needed to establish the functionality of scFv protein and the integration of

scFv-CDI gene into the plant genome and also improve transformation protocol in

banana cells.

90

CHAPTER 6: CONCLUSION

The anti-toxoplasmosis scFv gene (TOXO) ligated to CDI plant protease inhibitor gene

was putatively transformed into Nicotiana tabacum cv. BY-2 cell suspension using

pCTOXO-CDI-KDEL. In addition, the scFv anti-toxoplasmosis gene ligated to KDEL

fragment was also putatively transformed into tobacco cells using pCTOXO-KDEL by

Agrobacterium-mediated transformation method.

The effect of co-expression of plant protease inhibitor (CDI) on the expression of scFv

heterologous gene in plant cells was investigated by GUS fluorometric assay and

Bradford protein assay. The transient gene expression level was detected

fluorometrically in tissue extracts of tobacco cells and the TSP of transformed tobacco

cells was measured by Bradford assay.

The TSP of tobacco cells transformed with pCTOXO-CDI appeared to be higher than

tobacco cells transformed with pCTOXO-KDEL in Bradford protein assay. Therefore,

suggesting the positive effect of co-expression of Plant Protease Inhibitor (CDI) on the

expression of heterologous gene scFv anti-toxoplasmosis in tobacco cells.

91

6.1 FUTURE STUDIES

The results obtained from assessment of the GUS asaay analysis and total soluble

protein contents measurement are considered as preliminary findings for the real effects

of protease inhibitor in the transformed plant cells. Therefore, further research has to be

conducted to strength the results obtained.

The expression level of scFv anti-toxoplasmosis gene ligated to CDI and KDEL genes

was not verified in this research therefore, further studies need to be carried out to

determine the expression level of the recombinant protein using two different

constructs. The functionality of scFv anti-toxoplasmosis protein was also not studied

thus indicating the need for further research in this area.

92

REFERENCE:

1. Artsaenko, O., Kettig, B., Fiedler, U., Conrad, U. and During, K. (1998). Potato

tubers as a biofactory for recombinant antibodies. Molecular Breeding. 4: 313–319.

2. Benchabane, M., Goulet, C., Rivard, D., Faye, L., Gomord, V. and Michaud, D.

(2008). Preventing unintended proteolysis in plant protein biofactories. Plant

Biotechnology, 6: 633-648.

3. Birch-Machin, I., Newell, C.A., Hibberd, J.M. and Gray, J.C. (2004). Accumulation

of rotavirus VP6 protein in chloroplasts of transplastomic tobacco is limited by

protein stability. Plant Biotechnology Journal. 2: 261–270.

4. Bradford, M. (1976). A rapid and sensitive method for the quantitation of

microgram quantities of protein utilizing the principle of protein-dye binding.

Analytical Biochemistry. 72:248-254.

5. Callis, J. (1995). Regulation of protein degradation. Plant Cell, 7: 845–857.

6. Clark, D.P. (2005). Molecular Biology. Academic Press.600-607.

7. Compton, S.J. and Jones, C.G. (1985). Mechanism of dye response and interference

in the Bradford protein assay. Analytical Biochemistry. 151: 369-374.

8. Daniell, H., Streatfield, S.J. and Wyocoff, K. (2001). Medical molecular farming:

production of antibodies, biopharmaceuticals and edible vaccines in plants. Trends

in Plant science. 6: 219-226.

9. Daniell, H., Khan, M.S. and Allison, L. (2002). Milestones in chloroplast genetic

engineering: an environmentally friendly era in biotechnology. Trends Plant Sci. 7:

84–91.

10. Daniell H., Kumar S. and Dufourmantel N. (2005). Breakthrough in chloroplast

genetic engineering of agronomically important crops. Trends in Biotechnology, 23:

238-245.

93

11. Daniell, H. (2006). Production of biopharmaceuticals and vaccines in plants via the

chloroplast genome. Biotechnology. 1: 1071–1079.

12. Deneke, J., Botterman, J. and Deblaere, R. (1990). Protein secretion in plant cells

can occur via a default pathway. Plant Cell. 2: 51-59.

13. Doran, PM. (2006). Foreign protein degradation and instability in plants and plant

tissue cultures. Elsevier Trends in Biotechnology. vol.24 No.9.

14. Faye, L., Boulaflous, A., Benchabane, M., Gomord, V. and Michaud, D. (2005).

Protein modifications in the plant secretory pathway: current status and practical

implications in molecular pharming. Vaccine, 23: 1770-1778.

15. Fernandez-San Millan, A., Mingo-Castel, A., Miller, M. and Daniell, H. (2003). A

chloroplast transgenic approach to hyper-express and purify human serum albumin,

a protein highly susceptible to proteolytic degradation. Plant Biotechnology. 1: 71–

79.

16. Fiedler, U. and Conrad, U. (1995). High-level production and longterm storage of

engineered antibodies in transgenic tobacco seeds. Bio/Technology, 13: 1090–1093.

17. Fischer, R., Emans, N., Schuster, F., Hellwig, S. and Drossard, J. (1999). Towards

molecular farming in the future: using plant cell suspension cultures as bioreactors.

Biotechnol App Biochem. 30: 109-112.

18. Galili, G., Sengupta-Gopalan, C. and Ceriotti, A. (1998). The endoplasmic

reticulum of plant cells and its role in protein maturation and biogenesis of oil

bodies. Plant Molecular Biology. 38: 1–29.

19. Goulet, C. and Michaud, D. (2006). Degradation and stabilization of recombinant

proteins in plants. In: Floriculture, Ornamental and Plant Biotechnology, Vol. IV

(Taxeira da Silva, J., ed.), pp. 35–40. London: Global Science Books.

20. Gupta, S. Codon Optimization. (2003). Retrieved December 20, 2010, from

http://www.guptalab.org/shubhg/pdf/shubhra_codon.pdf.

94

21. Hajdukiewicz, P., Svab, Z., Maliga, P., (1994). The small versatile pPZP family of

Agrobacterium binary vectors for plant transformation. Plant Mol Biol 25:989-994

22. Hellwig, S., Drossard, J., Twyman, RM. and Fischer, R. (2004). Plant cell cultures

for the production of recombinant proteins. Nature Biotechnol, 22: 1415-1422.

23. Hendy, S., Chen, ZC., Barker, H., Santa Cruz, S., Chapman, S., Torrance, L.,

Cockburn, W., Whitelam, GC. (1999). Rapid production of single-chain Fv

fragments in plants using a potato virus X episomal vector. Journal Immunol

Methods. 231: 137-146

24. Hiatt, A., Cafferkey, R. and Bowdish, K. (1989). Production of antibodies in

transgenic plants. Nature, 342: 76-8.

25. Hyunjong, B., Lee, D.S. and Hwang, I. (2006) Dual targeting of xylanase to

chloroplasts and peroxisomes as a means to increase protein accumulation in plant

cells. Journal of Experimental Botany. 57: 161–169.

26. Jefferson, R.A., Kavanagh, T.A. and Bevan, M.W. (1987). GUS fusions: B-

glucuronidase as a sensitive and versatile gene fusion marker in higher plants.

EMBO Journal. 6: 3901-3907.

27. Jeon, G.A., Eum, J.S. and Sim, W.S. (1998). The role of inverted repeat (IR)

sequence of the virE gene _expression in Agrobacterium tumefaciens pTiA6.

Molecules and Cells, 8: 49-53.

28. Joosten, V., Lokman, C., Cees A.M.J.J Van Den Hondel and Punt, PJ. (2003). The

production of antibody fragments and antibody fusion proteins by yeasts and

filamentous fungi. Microbial cell factories, 2: 1-7.

29. Jung, E. and Williams, KL. (1997). The production of recombinant glycoproteins

with special reference to simple eukaryotes including Dictyostelium dicoideum.

Biotechnol. Appl. Biochem. 25: 3-10.

95

30. Kato, Y., Murakami, S., Yamamoto, Y., Chatani, H., Kondo, Y., Nakano, T.,

Yokota, A. and Sato, F. (2004). The DNA-binding protease, CND41, and the

degradation of ribulose-1,5-biphosphate carboxylase/oxygenase in senescent leaves

of tobacco. Planta, 220: 97–104.

31. Kato, Y., Yamamoto, Y., Murakami, S. and Sato, F. (2005) Posttranslational

regulation of CND41 protease activity in senescent tobacco leaves. Planta, 222:

643–651.

32. Komarnytsky, S., Borisjuk, N., Yakoby, N., Garvey, A. and Raskin, I. (2006).

Cosecretion of protease inhibitor stabilizes antibodies produced by plant roots. Plant

Phisiology, 141: 1185-1193.

33. Kusnadi, A.R., Nikolov, Z.L. and Howard, J.A. (1997). Production of recombinant

proteins in transgenic plants: Practical considerations. Biotechnology and

Bioengineering. 56: 473-484.

34. Lelivelt C.L., McCabe M.S., Newell C.A., Desnoo C.B., van Dun K.M., Birch-

Machin I., Gray J.C., Mills K.H. and Nugent J.M. (2005). Stable plastid

transformation in lettuce (Lactuca sativa L.) Plant Molecular Biology, 58: 763-774.

35. Lin, J.F. and Wu, S.H. (2004). Molecular events in senescing Arabidopsis leaves.

Plant Journal. 39: 612–628

36. Lu, Y., Wu, X., Xu, Z., Li, Q. and Deng, X. (2004). Advances in Dictyostelium

discoideum as an expression system. Chemistry Journal, 6: 58-68.

37. Ma, J., Hiatt, A., Hain, M., Vine, N.D., Wang, F., Stabila, P., van Dolleweerd, C.,

Mostov, K. and Lherner, T. (1995). Generation and assembly of secretory antibodies

in plants. Nat. Rev. Genet. 268: 716-719.

38. Ma, J.K., Drake, P.M. and Christou, P. (2003). The production of recombinant

pharmaceutical proteins in plants. Nature Reviews Genetics. 4: 794–805.

96

39. Mackenzie, S.A. (2005). Plant organellar protein targeting: a traffic plan still under

construction. Trends Cell Biology. 15: 548–554.

40. Michaud, D., Anguenot, R. and Brunelle, F. (2005). Method for increasing protein

content in plant cells. US Patent application, 2005/0055746.

41. Nagata, T., Nemoto, Y., Hasezawa, S. (1992). Tobacco BY-2 cell line as the "HeLa"

cell in the cell biology of higher plants. International Review of Cytology. 132: 1-

30.

42. Nouaille., Luciana. AR., Anderson, M., Pontes, D., Le Loir, Y., Sergio, CO.,

Phillippe, L. and Vasco, A. (2003). Heterologous protein production and delivery

systems for Lactococcus lactis. Genet. Mol. Res., 2: 102-111.

43. Nugent G.D., Coyne S., Nguyen T.T. and Kavanagh T.A., Dix P.J .(2006). Nuclear

and plastid transformation of Brassica oleracea var. botrytis (cauliflower) using

PEG-mediated uptake of DNA into protoplasts. Plant Science. 170: 135-142.

44. Nuttall, J., Vine, N., Hadlington, J.L., Drake, P., Frigerio, L. and Ma, J.K. (2002).

ER-resident chaperone interactions with recombinant antibodies in transgenic

plants. Eur. J. Biochemistry. 269: 6042–6051.

45. Opabode, J.T. (2002). Factors influencing transformation of crops by

Agrobacterium tumefaciens. A review seminar presented at Department of Plant

Science, Obafemi Awolowo University, Nigeria, 23 March 2002.

46. Opabode, J.T. (2006). Agrobacterium-mediated transformation of plants: emerging

factors that influence efficiency. Biotechnology and Molecular Biology Review. 1:

12-20.

47. Otegui, M.S., Noh, Y.S., Martinez, D.E., Vila Petroff, M.G., Staehlin, L.A.,

Amasino, R.M. and Guiamet, J.J. (2005). Senescence associated vacuoles with

intense proteolytic activity develop in leaves of Arabidopsis and soybean. Plant

Journal. 41: 831–844.

97

48. Outchkourov, N.S., Rojelj, B., StrukelJ, B. and Jongsma, M.A. (2003). Expression

of sea anemone equistatin in potato. Effects of plant proteases on heterologous

protein production. Plant physiology, 133: 379-390.

49. Parrott, D., Yang, L., Shama, L. and Fischer, A.M. (2005). Senescence is

accelerated and several proteases are induced by carbon ‘feast’ conditions on barley

(Hordeum vulgare L.) leaves. Planta, 222: 989–1000.

50. Pagny, S., Cabanes-Macheteau, M., Gillikin, J.W., Leborgne-Castel, N., Lerouge,

P., Boston, R.S., Faye, L. and Gomord, V. (2000). Protein recycling from the golgi

apparatus to the endoplasmic reticulum in plants and its minor contribution to

calreticulin retention. Plant Cell, 12: 739–756.

51. Park, M., Kim, S.J., Vitale, A. and Hwang, I. (2004). Identification of the protein

storage vacuole and protein targeting to the vacuole in leaf cells of three plant

species. Plant Physiology. 134: 625–639.

52. Rai, M. and Padh, H. (2001). Expression systems for production of heterologous

proteins. Current Science, 80: 1121-1128.

53. Richter, L.J., Thanavala, Y., Arntzen, C.J. and Mason, H.S. (2000). Production of

Hepatitis B surface antigen in transgenic plants for oral immunization. Nature

Biotechnology, 18: 1167-1171.

54. Rivard, D., Anguenot, R., Brunelle, F., Le, V.Q., Vezina, L.P., Trepanier, S. and

Michaud, D. (2006). An in-built proteinase inhibitor system for the protection of

recombinant proteins recovered from transgenic plants. Plant Biotechnology, 4: 359-

368.

55. Robinson, D.G., Oliviusson, P. and Hinz, G. (2005). Protein sorting to the storage

vacuoles of plants: a critical appraisal. Traffic, 6: 615–625.

56. Ryan, K.J. and Ray, C.G. (editors) (2004). Sherris Medical Microbiology (4th ed.).

McGraw Hill. pp. 723–7.

98

57. Schaller, A. (2004) A cut above the rest: the regulatory function of plant proteases.

Planta, 220: 183–197.

58. Sharp, J.M. Doran, P.M. (2001). Characterization of monoclonal antibody fragments

produced by plant cells. Biotechnology and Bioengineering. 73: 338-346.

59. Shewry, P.R. and Halford, N.G. (2002). Cereal seed storage proteins: structures,

properties and role in grain utilization. Journal of Experimental Botany. 53: 947–

958.

60. Slater, A., Scott, N. and Fowler, M. (2003). Plant biotechnology: The genetic

manipulation of plants. New York: Oxford University Press. pp. 55-58.

61. Smith, M.L., Keegan, M.E., Mason, H.S. and Shuler, M.L. (2002). Factors

Important in the Extraction, Stability and in vitro Assembly of the Hepatitis B

Surface Antigen Derived from Recombinant Plant Systems. Biotechnology

Progress, 18: 538-550.

62. Stein, K.E. Webber, K.O. (2001). The regulation of biologic products derived from

bioengineered plants. Current opinion in biotechnology. 12: 308-311.

63. Stevens, L.H., Stoopen, G.M., Elbers, I.J.W., Molthoff, J.W., Bakker, H.A.C.,

Lommen, A.,Bosch, D. and Jordi, W. (2000). Effect of climate conditions and plant

developmental stage on the stability of antibodies expressed in transgenic tobacco.

Plant Phisiology. 124: 173-182.

64. Stoger, E., Vaquero, C., Torres, E., Sack, M., Nicholson, L., Drossard, J., Williams,

S., Keen, D., Perrin, Y., Christou, P. and Fischer, R. (2000). Cereal crops as viable

production and storage systems for pharmaceutical scFv antibodies. Plant Molecular

Biology. 42: 583–590.

65. Stoger, E., Ma, J.K., Fischer, R. and Christou, P. (2005). Sowing the seeds of

success: pharmaceutical proteins from plants. Curr. Opin. Biotechnology. 16: 167–

173.

99

66. Torda, A. (2001). Toxoplasmosis. Are cats really the source?. Australian Family

Physician, 30 (8): 743–7.

67. Torres, E., Vaquero C., Nicholson, L., Sack, M., Stoger, E., Drossard, J., Christou,

P., Fischer, R. and Perrin, Y. (1999). Rice cell culture as an alternative production

system for functional diagnostic and therapeutic antibodies. Transgenic Res. 8: 441-

449.

68. Twyman, RM., Schillberg, S. and Fischer, R. (2007). P.Ranalli (editor).

Improvement of Crop Plants for Industrial End Uses: Molecular farming of

antibodies in plants. Springer, 435-469.

69. van der Vyver, C., Schneidereit, J., Driscoll, S., Turner, J., Kunert, K. and Foyer,

C.H. (2003). Oryzacystatin I expression in transformed tobacco produces a

conditional growth phenotype and enhances chilling tolerance. Plant Biotechnology.

1: 101–112.

70. Vierstra, R.D. (2003) The ubiquitin/26S proteasome pathway, the complex last

chapter in the life of many plant proteins. Trends Plant Sci. 8: 135–142.

71. Vitale, A. and Hinz, G. (2005). Sorting of proteins to storage vacuoles: How many

mechanisms? Trends Plant Science. 10: 316–323.

72. Wandelt, C.I., Khan, M.R., Craig, S., Schroeder, H.E., Spencer, D. and Higgins, T.J.

(1992). Vicilin with carboxy-terminal KDEL is retained in the endoplasmic

reticulum and accumulates to high levels in the leaves of transgenic plants. Plant

Journal. 2: 181–192.

73. Watson, J.M., Fusaro, A.F., Wang, M. and Waterhouse, P.M. (2005). RNA silencing

platforms in plants. FEBS Letters. 579: 5982–5987.

74. Whitelam, GC. and Cockburn, W. (1997). Plantibodies: antibodies produced in

transgenic plants. The Biochemist, 19: 7-9.

100

75. Yang, J., Barr, L.A., Fahnestock, S.R. and Liu, Z. (2005). High yield recombinant

silk-like protein production in transgenic plants through protein targeting.

Transgenic research, 14: 313-324.

76. Yazaki, PJ. and Wu AM. (2004). Expression of recombinant antibodies in

mammalian cell lines. Methods Mol Biol, 248: 255-68.

77. Zupan, J., Muth, T.R, Draper, O. and Zambryski, P. (2000). The transfer of DNA

from Agrobacterium tumefaciens into plants: A feast of fundamental insights. The

Plant Journal, 23: 11-28.

78. pCambia vectors. Retrieved September 7,2010, from

http://www.cambia.org/daisy/cambia/585.html

101

APPENDICES

102

APPENDIX - A

103

104

105

106

107

108

109

110

111

112

113

114

APPENDIX - B

FORWARD DNA SEQUENCING RESULT ANALYSIS

1. Orange sequence matches the scFv gene

TCCACTATTTAACTAGCCTGCAGAGAAGTGACCAGAGTCCCTTGGCC

CCAGTAAGCAAACCAGGCCGTGGCCGTAGTAAGTCCTCGACTTGCA

CAGTAATAGACGGCAGAATCCTCAGATGTCAGGCTGCTGAGTTGCAT

GTAGGCTGTGTTGGAGGATGTATCTGCAGTGAATGCGGCCTTGCCCT

TGAAGTTCTCATAGTAGTTAGAACTACCACTTCCAGGATAAATCTCTC

CAATCCACTCAAGGCCATGTCCAGGCCTCTGCTTTACCCACTCTATC

CAGTAGCTACTGAATGTGTAGCCAGTAGCCTTGCAGGATATCTTCAC

TGAGGCCCCAGGCCTCACCAGCTCAGCTCCAGACTCCACCCCCTGC

ACCTCGAGGGAAGATCTAGAGGAACCACCTTTCAGCTCCAGTTGGTC

CCAGCACCGAACGTGAGCGGAGTACTATAATGTTGCTGACAGTAATA

AACTGCCAGGTCTTCAGCCTGCACACTGCTGATGGTGAAAGTGAAAT

CCGTCCCAGATCCACTGCCAGTGAAGCGATCAGGGACTCCAGTGTA

CCGGTAGGATGCCGAGTAAATCAGTAGTTTAGGAGATTGTCCGGGTT

TCTGTTGATACCAGGCTACAGCAGTACTCACATCCTGACTGGCCTTG

CAGGTGATGCTGACCCTGTCTCCTACTGATGTGGACATGAATTTGTG

AGACTGTGTCAACACAATGTCGAGCTCGGCCGGCTGGGCCGCATAG

AAAGGAACGGATCTGAGGGTAAATTTCTAGTTTTTCTCC

115

APPENDIX - C

REVERSE DNA SEQUENCING RESULT ANALYSIS

1. The row of N at the 5’ end of the sequence shows the sequencing error. No

specific nucleotide was detected.

2. Orange sequence matches the scFv gene.

NNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNNN

NNNNNNNNNNNNNNNTTTNNTTTTTTTNNNNAAACANGGGACTCTTG

ANCATGGTTGGNTGCAGNGACAGTGACCAGAGTCCCTTGGCCCCAG

TAAGCAAACCAGGCCGTGGCCGTAGTAAGTNCTCGACTTGCACAGTA

ATAGACGGCAGAATCCTCAGATGTCAGGCTGCTGAGTTGCATGTAGG

CTGTGTTGGAGGATGTATNNGCAGTGAATGCGGCCTTNCCCTNGAAG

TTCTCATAGTAGTTAGAACTACCACTTCCAGGATAAATCTNTNCAATC

CACTCAAGGCCATGTCCAGGCCTCTGCTTTACCCACTCTATCCAGTA

GNTACTGAATGTGTAGCCAGTAGCCTTGCAGGATATCTTCANTGAGG

CCCCAGGCCTCACCAGCTCAGCTCCAGACTCCACCCCCTGCACCTC

GAGGGAAGATCTAGAGGAACCACCTTTCAGCTCCAGTTGGTCCCAGC

ACCGAACGTGAGCGGAGTACTATAATGTTGCTGACAGTAATAAACTG

CCAGGTCTTCAGCCTGCACACTGCTGATGGTGAAAGTGAAATCCGTC

CCAGATCCACTGCCAGTGAAGCGATCAGGGACTCCAGTGTACCGGT

AGGATGCCGAGTAAATCAGTAGTTTAGGAGATTGTCCTGGTTTCTGTT

GATACCAGGNTACAGCAGTACTCACATCCTGACTGGCCTTGCAGGNG

ATGCTGACCCTGTCTCCTACTGATGTGGACATGAATTTGTGAGANCT

GTGTCAACACAATGTCCGAGGCTCGGTCCGGNTGGGCCCGCATAGA

AAGGAANCGGATTTGACGGCAAANGN

116

APPENDIX - D

ANALYSIS OF COMBINED FORWARD AND REVERSE DNA SEQUENCING

RESULT

1. Red sequence matches the pCambia1301 sequence (GenBank: AF234297.1)

2. Blue sequence is BamHI (ggatcc) sequence which is located at 3’ end of scFv

gene sequence.

3. Green and orange sequence smatche the scFv gene.

4. PI gene (BBTI) sequence is missing in the sequencing result.

CTTGANCATGGTTGGNTGCAGNGACAGTGACCAGAGTCCCTTGGCCCCA

GTAAGCAAACCAGGCCGTGGCCGTAGTAAGTNCTCGACTTGCACAGTAA

TAGACGGCAGAATCCTCAGATGTCAGGCTGCTGAGTTGCATGTAGGCTG

TGTTGGAGGATGTATCTGCAGTGAATGCGGCCTTGCCCTTGAAGTTCTC

ATAGTAGTTAGAACTACCACTTCCAGGATAAATCTCTCCAATCCACTCAA

GGCCATGTCCAGGCCTCTGCTTTACCCACTCTATCCAGTAGCTACTGAAT

GTGTAGCCAGTAGCCTTGCAGGATATCTTCACTGAGGCCCCAGGCCTCA

CCAGCTCAGCTCCAGACTCCACCCCCTGCACCTCGAGGGAAGATCTAGA

GGAACCACCTTTCAGCTCCAGTTGGTCCCAGCACCGAACGTGAGCGGA

GTACTATAATGTTGCTGACAGTAATAAACTGCCAGGTCTTCAGCCTGCAC

ACTGCTGATGGTGAAAGTGAAATCCGTCCCAGATCCACTGCCAGTGAAG

CGATCAGGGACTCCAGTGTACCGGTAGGATGCCGAGTAAATCAGTAGTT

TAGGAGATTGTCCGGGTTTCTGTTGATACCAGGCTACAGCAGTACTCACA

TCCTGACTGGCCTTGCAGGTGATGCTGACCCTGTCTCCTACTGATGTGG

ACATGAATTTGTGAGACTGTGTCAACACAATGTCGAGCTCGGCCGGCTG

GGCCGCATAGAAAGGAACGGATCTGAGGGTAAATTTCTAGTTTTTCTCC

117

APPENDIX - E

SCFV GENE SEQUENCE

5’NNNNACACTTTCACAGTCTATGCGGCACGCGGTTCCAGCGGATCCGGATA

CGGCACCGGCGCACCTGCGGCCGCCCACTAGTGACAGATGGGGCTGTTGTT

TTGGCTGCAGAGACAGTGACCAGAGTCCCTTGGCCCCAGTAAGTCCGTCCC

AGTTGTGCACAGTAATAGACTGCAGAGTCCTCAGATGTCAGGCTGCTGAGC

TGCATGTAGGCTGTGTTGGAGGATTTGTCTACATTCAATGTGGCCTTGTCCT

TGAACTTCTGATTTAACCTAGTTTCACTATTGGAAGGATCAATCATGCCAAT

CCACTCAAGGCCTTGTCCAGGCCTCTGTTTCACCCAGTGCATCCAGTAGCTG

GTGAAGGTATAGCCTGAAGCCTTGCAGGACATCTTCACTGAAGCCCAGGCC

TCACCAGCTCAGGCCCAGACTCCACCAGCTGCACCTCGAGGGAAGATCTAG

AGGAACCACCTTTGATTTCCAGCTTGGTCCCCCCTCCGAACGTGTACGGAAT

CTCCCAACTGTGCTGACAGTAATATGTTGCAGTATCCTCCTCCTCCACAGGA

TGGATGTTGAGGGTGAAGTCTGTCCCAGACCCACTGCCACTGAACCTGGCA

GGGACCCCAGATTCTAGGTTGGATGCATACTTGATGAGGAGTTTGGGTGGC

TGTCCTGGTTTCTGTTGGTACCAGTGCATATAACTATAGCTAGATGTACTGA

CACTTTGGCTGGCCCTGCATGAGATGGTGGCCCTCTGCCCCAGAGATACAGC

TAAGGAAGCAGGAGACTGTGTCATCACAATGTCGAGCTCGGCCGGCTGGGC

CGCATAGAAAGGAACAACTAAAGGAATTGCGAATAATAATTTTTTCACGTT

GAAAATCTCCAAAAAAAAGGCTCCAAAGCTTGGCGTAATCATGGTCATAGC

TGTTTCCTGTGTGAAATTGTTATCCGCTCACAATTCCACACAACATACGAGC

CGGAAGCATAAAGTGTAAAGCCTGGGGTGCCTAATGAGTGAGCTAACTCAC

ATTAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTCGGGC

CACCTGCATTAATGAATCGGCCAACCCCGCGGGGAAAGGCGGTTTGCGTAT

TGGGCGCTCTTTCCGCTTCCTCGCCAATGAACCCGTGCCCTCGGTCGTTCGC

118

CTGCCGGCGAAAGCGGTTAATCCAGCCTCACCTCAGAGGCGGTTATNNNGG

TTATTCNACCAAGAANNNNNGGGGNNTAACCCGCGGAGAGNANGAANNAN

TTGTGTTGN-3’

119

APPENDIX - F

BBTI GENE SEQUENCE

5’ggatcctacagcgtgcaagcaccgcagaaacaaacaaaaaaacacccgggaaatccaagcagctgttgcaaccggttcc

ttcgaggagaacgtgagaaagaagcatgaggccccaggtgctgctcgtcgcactggccgtcttcgccgtcctcgtagctctg

ccactgggcaaagcgcacgaggaggaggaggaaggggttgaactcgaagccagcaggaggcggtggccgtgctgcgac

cagtgcggcatttgcaccaggtcgcagccgccgatatgcgagtgcagggacacgtcgacgaccggctgccacccggcttg

caaggcgtgcgccctgtccatctccgacggcctcttcgtgtgcaaggacaagatcgtcaacttctgcaagcgccgctgcaccc

gtcgtactgatgatgatgatgcgtgacttataatcacttaattaggcaaataaccttgccaataaataatttccgttcggagctccat

ttgcatctttctcaagatgcagacgacagggtttaaataatcaaggcgatgcttgcttgctaaaaaaaaaaaaatctgagaaaga

tgagctctagagatct-3’

120

APPENDIX - G

CDI GENE SEQUENCE

5’ggatcctacaatataccaatcaatatgatgaagtgtttatttttgttatgtctgtgtttgtttcccattgtggtgttttcatcaagtttca

cttcccaaaatcccattgaattaccgagtgcttctcctaaacctaatccagtacttgacacaaatggtaacgaacttaatcctaattc

gagctatcgcattatttccacattttggggtgcgttaggtggtgatgtatacctaggtaaatctccaagatcatctgccccttgtcta

gatggtgtgtttcgttacaattctgatgttggaactgtcggtacacccgttagattcattcctttatctggaggtatatttgaagatca

actaatggacctacaattcaacattgcaacagtgaaattgtgtgttagttatacaatttggaaagccggtaatttaaatgcatactat

agggcgatgttgttggagacggaaggaagcataggacaagtagatagcagttatttcaagattgttaaagcatcaacatttggtt

acaacttactctattgccctattactcgtcctgtgctttgtccattttgccgtggtgatggcttctgtgcaaaagtgggtgtaattaatc

aagatggaagaaggcgtttggctcttgtcaacgaaaatcctcttggggtctacttcaagaaagtctagtatgtttatgctatgttat

gttatgttgtaaaataaacacatgctaacgtatatctatatattttagcatgtctttcttaataaaattggagtgtttgcttatataaaaaa

aaaaaaaaatctgagaaagatgagctctagagatct-3’

121

APPENDIX - H

OCPI GENE SEQUENCE

5’ggatcctacgcattcgctagccacgccgtccgctcaggccgaggcgcatcgcgcagggggagaaggggaggagaaga

tgtcgagcgacggagggccggtgcttggcggcgtcgagccggtggggaacgagaacgacctccacctcgtcgacctcgcc

cgcttcgccgtcaccgagcacaacaagaaggccaattctctgctggagttcgagaagcttgtgagtgtgaagcagcaagttgt

cgctggcactttgtactatttcacaattgaggtgaaggaaggggatgccaagaagctctatgaagctaaggtctgggagaaac

catggatggacttcaaggagctccaggagttcaagcctgtcgatgccagtgcaaatgcctaaggcccatctcgtatcctatgtg

tatcaagttatcaagaagatggggaataatatggtgtggatatagctattggacatgttaattatccacatgataatatggcttggat

ataaggatctcacacgataatatggcttggatatatagctattaaagattttacctatggcatatttcaatgtgtattagtactaagta

agaatgattgcaaggtgtattaactacaaatattgcaataaaagtccctgttactacaacttacaaggtctgagaaagatgagctc

tagagatct-3’

122

APPENDIX - I

4-MU STANDARD MEASUREMENTS

0.2M

Carbonate

buffer (ml)

1µM 4-

MU stock

(µl)

Sample

conc.(X)

(nM)

Fluorescence

Reading

replicate1

Fluorescence

Reading

replicate2

Average

2.0 0 0 0 0 0

1.9 100 50 250 240 245

1.8 200 100 600 598 599

1.7 300 150 905 894 899.5

1.6 400 200 1210 1200 1205

123

APPENDIX - J

4-MU STANDARD CURVE

y = 6.129x - 23.2 R² = 0.9978

-200

0

200

400

600

800

1000

1200

1400

0 50 100 150 200 250

Fluorescent

Intensity

4-MU (pmol)

Standard Dilution of

4-Methylumbelliferone and

Relative Fluorescent Intensity

124

APPENDIX - K

OD VALUES FOR DIFFERENT BSA CONCENTRATIONS IN PROTEIN

STANDARD CURVE ASSAY

BSA conc.

(mg)

OD replicate1 OD replicate3 OD replicate 3 Average

0 0.537 0.534 0.529 0.533

0.0025 0.577 0.581 0.585 0.581

0.005 0.617 0.619 0.615 0.617

0.0075 0.643 0.639 0.642 0.641

0.01 0.645 0.647 0.643 0.645

0.015 0.652 0.649 0.647 0.649

0.02 0.675 0.668 0.671 0.671

0.025 0.725 0.736 0.728 0.729

0.03 0.793 0.799 0.785 0.792

125

APPENDIX - L

BRADFORD STANDARD CURVE

y = 7.0074x + 0.5614 R² = 0.9169

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

0 0.01 0.02 0.03 0.04

Standard Protein curve

A595

BSA protein CONCENTRATION (mg)

126

APPENDIX - M

DNA LADDER 100BP

127

DNA LADDER 1 KB